WO2006039285A2 - Compositions and methods for inducing adipose tissue cell death - Google Patents

Compositions and methods for inducing adipose tissue cell death Download PDF

Info

Publication number
WO2006039285A2
WO2006039285A2 PCT/US2005/034632 US2005034632W WO2006039285A2 WO 2006039285 A2 WO2006039285 A2 WO 2006039285A2 US 2005034632 W US2005034632 W US 2005034632W WO 2006039285 A2 WO2006039285 A2 WO 2006039285A2
Authority
WO
WIPO (PCT)
Prior art keywords
derivatives
precursors
ajoene
garlic extract
pharmaceutical composition
Prior art date
Application number
PCT/US2005/034632
Other languages
French (fr)
Other versions
WO2006039285A3 (en
Inventor
Changlong Li
Maryanne Dellafera
Clifton A. Baile
Original Assignee
The University Of Georgia Research Foundation, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The University Of Georgia Research Foundation, Inc. filed Critical The University Of Georgia Research Foundation, Inc.
Publication of WO2006039285A2 publication Critical patent/WO2006039285A2/en
Publication of WO2006039285A3 publication Critical patent/WO2006039285A3/en

Links

Classifications

    • 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
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/88Liliopsida (monocotyledons)
    • A61K36/896Liliaceae (Lily family), e.g. daylily, plantain lily, Hyacinth or narcissus
    • A61K36/8962Allium, e.g. garden onion, leek, garlic or chives

Definitions

  • compositions and Methods for Inducing Adipose Tissue Cell Death which is incorporated herein by reference in its entirety.
  • the present disclosure is generally related to compositions and methods for
  • compositions administration of the compositions to hosts and, more particularly, is related to
  • compositions designed for treatment for inducing adipose tissue cell death and
  • Obesity represents a major public health issue that continues to grow, accounts
  • Obesity is marked by excess adipose (i.e., fat) tissue
  • diabetes e.g., decreased insulin sensitivity
  • liposuction is an invasive and painful
  • osteoporosis a disease that results in over 1.5 million bone fractures a year.
  • An illustrative embodiment of a method of treating obesity is disclosed.
  • the compound is present in a dosage level effective to initiate the release or
  • the apoptosis inducing factor leads to the
  • tissue cells in a host includes initiating the release or activation of an
  • the extract compound The apoptosis inducing factor leads to apoptotic cell death.
  • the at least one garlic extract compound is present in a dosage
  • the apoptosis inducing factor leads to the apoptotic cell death of adipose tissue cells in a host.
  • composition including at least one garlic extract compound in combination with a pharmaceutically acceptable carrier.
  • the garlic extract compound in combination with a pharmaceutically acceptable carrier.
  • osteoporosis is present in a dosage level effective to treat osteoporosis.
  • FIG. 1 illustrates the effect of ajoene on cell viability. 3T3-L1 adipocytes were
  • FIGS. 2A and 2B illustrate the effect of ajoene on apoptosis.
  • FIG. 2A 3T3-
  • Ll adipocytes were treated with ajoene at various concentrations (0, 10, 50, 200, 400
  • FIG. 2B illustrates apoptosis by gel
  • 3T3-L1 adipocytes were incubated with ajoene at various
  • Lane 1 analyzed by gel electrophoresis; Lane 2, 100 ⁇ M ajoene; Lane 3, 200
  • FIG. 3 A and 3B illustrate the effects of ajoene on intracellular hydrogen
  • FIG. 3 A illustrates the time course of the effect of ajoene on
  • 3T3-L1 adipocytes were grown in 96-well plates and treated with
  • FIG. 3B illustrates the reduction of ajoene-induced apoptosis in cells pretreated with NAC. The percentage of apoptotic cells measured
  • FIGS. 4A-4D illustrate the effect of ajoene on MAPKs phosphorylation.
  • the protein levels of unphosphorylated and phosphorylated forms of MAP kinases were
  • ERKl/2 and JNK were performed. Integrated density values (phosphorylated/total)
  • FIGS. 5A-5C illustrate the effect of ajoene on PARP cleavage and caspase3
  • FIG. 5A 3T3-L1 adipocytes were treated with ajoene (200 ⁇ M) for the
  • ⁇ -Actin lower panel was used as an equal loading control.
  • FIG. 5B 3T3-L1 adipocytes were preincubated with 10 mM NAC for 1 hr
  • FIGS. 6A-6B illustrate ajoene induced translocation of AIF from mitochondria
  • FIG. 6A 3T3-L1 adipocytes were treated with ajoene (200 ⁇ M) for 0, 1, 4, 8 and 12 hr. Equal amounts of AIF protein from nuclear fraction (upper panel) and
  • Integrated density values (AIF/actin) were calculated and expressed as % highest
  • mitochondrial means with different letters are different across time: xyz: p ⁇ 0.01.
  • FIG. 6B 3T3-L1 adipocytes were preincubated with 10 mM NAC for 1 hr in
  • organism or "host” refers to any living entity comprised of at least
  • a living organism can be as simple as, for example, a single eukaryotic cell
  • “host” includes humans, mammals (e.g., cats, dogs, horses, chicken, pigs, hogs, cows,
  • host includes humans, companion animals (e.g., cats, dogs, and the like),
  • apoptosis inducing factor refers to any compound
  • apoptosis inducing factor activated in a pathway leading to apoptosis.
  • apoptosis inducing factor activated in a pathway leading to apoptosis.
  • AIF Apoptosis Inducing Factor protein
  • apoptosis inducing factors in the present disclosure include,
  • ROS reactive oxygen species
  • MAPKs mitogen-activated protein kinases
  • PARPs PoIy(ADP ribose) polymerases
  • terapéuticaally effective amount refers to that
  • therapeutically effective amount refers to that amount that has the effect of (1) causing
  • apoptosis of adipose cells and/or (2) reducing the mass of the adipose cells/tissue.
  • “Pharmaceutically acceptable salt” refers to those salts that retain the
  • hydrobromic acid sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid,
  • ethanesulfonic acid p-toluenesulfonic acid
  • salicylic acid malic acid, maleic acid
  • esters as used herein refers to those wherein
  • esters of one or more compounds of the composition that, within the scope of sound
  • a “pharmaceutical composition” refers to a mixture of one or more of the
  • composition One purpose of a pharmaceutical composition is to facilitate
  • a "pharmaceutically acceptable carrier” refers to a carrier or diluent that does not cause significant irritation to an organism and does not abrogate
  • excipient refers to an inert substance added to a pharmaceutical
  • composition to further facilitate administration of a compound examples include:
  • excipients include, without limitation, calcium carbonate, calcium phosphate, various combinations thereof
  • Treating" or “treatment” of a condition includes preventing the condition
  • prodrug refers to an agent that is converted into a biologically
  • Prodrugs are often useful because, in some situations, they may
  • the prodrug may also have improved solubility in pharmaceutical compositions over the parent
  • a prodrug may be converted into the parent drug by various mechanisms,
  • topically active agents refers to compositions of the
  • topically refers to application of the compositions
  • salt(s) as used herein.
  • Pharmaceutically acceptable ⁇ i.e., non-toxic, physiologically
  • salts are preferred, although other salts are also useful ⁇ e.g., in isolation or
  • compounds may be formed, for example, by reacting the compound with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt
  • the disclosed compounds that contain a basic moiety may form salts with a
  • Exemplary acid addition salts include acetates
  • benzenesulfonates bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates,
  • methanesulfonates formed with methanesulfonic acid
  • 2-naphthalenesulfonates formed with 2-naphthalenesulfonates
  • nicotinates such as nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as
  • tartrates tartrates, thiocyanates, toluenesulfonates (such as tosylates), undecanoates, and the
  • the disclosed compounds that contain an acidic moiety may form salts with a
  • exemplary basic salts include ammonium
  • alkali metal salts such as sodium, lithium, and potassium salts
  • alkaline earth salts such as sodium, lithium, and potassium salts
  • metal salts such as calcium and magnesium salts; salts with organic bases (for
  • organic amines such as benzathines, dicyclohexylamines, hydrabamines
  • Basic nitrogen-containing groups may be quaternized with agents such as
  • lower alkyl halides e.g., methyl, ethyl, propyl, and butyl chlorides, bromides and
  • dialkyl sulfates e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates
  • chain halides e.g., decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides
  • aralkyl halides e.g., benzyl and phenethyl bromides
  • Solvates of the compounds are also contemplated herein. Solvates of the compounds are preferably hydrates.
  • isomers or may be admixed, for example, as racemates or with all other, or other
  • compositions for inducing adipose tissue cell death, and methods for treating
  • compositions include, but are not limited to, compositions having at least one garlic extract compound and, in particular, garlic thiosulfinates and
  • the method includes inducing adipose
  • tissue cell death in a host by administering a composition having at least one garlic
  • the methods include treating conditions such as, but not
  • compositions having at least one
  • Obesity is a chronic and costly condition that is increasing rapidly throughout the world. Obesity is considered a major risk factor for noninsulin-dependent diabetes
  • Adipose tissue mass is determined by processes governing adipocyte size and number (3).
  • Reduction of adipocyte number can result from preadipocyte and adipocyte apoptosis
  • apoptosis may be an important
  • the garlic extract compound of the present disclosure e.g., ajoene
  • apoptosis inducing factor leads to the apoptotic cell death of adipose tissue cells.
  • This apoptotic cell death is unexpectedly caspase-independent, which is in
  • apoptosis refers to a physiological process wherein selected cells
  • Apoptosis involves a sequence of
  • ROS are recognized to play a key role in cell signaling.
  • the cellular level
  • oxidant injury elicits a broad spectrum of responses ranging from proliferation to growth arrest, to senescence, to cell death (for review see 9).
  • oxidant injury elicits a broad spectrum of responses ranging from proliferation to growth arrest, to senescence, to cell death (for review see 9).
  • MAPK activated protein kinases
  • ERKl/2 Jun-N-teraiinal kinase
  • JNK Jun-N-teraiinal kinase
  • JNK and p38 MAPK activation is associated with apoptosis induction, whereas ERK activation is cytoprotective (14).
  • PARP which contributes to the pathogenesis of various diseases (15, 16).
  • Apoptosis Inducing Factor was more recently cloned and identified as a
  • ATF mitochondrial intermembrane space protein. In response to apoptotic stimuli, ATF is
  • adipocytes is initiated by the generation of hydrogen peroxide, which leads to
  • ROS reactive oxygen species
  • MAPK protein kinases
  • ERK extracellular signal-regulating kinase 1/2
  • c-Jun-N protein kinases
  • JNK N-terminal kinase
  • PARP polymerase
  • ROS reactive oxygen species
  • N-acetyl- L -cysteine NAC
  • Annexin V (AV)
  • FITC-conjugated AV is
  • chromosomal DNA to be exposed.
  • Propidium iodide a fluorescent dye that binds to
  • DNA can be used in conjunction with FITC-conjugated AV to identify subpopulations
  • the TUNEL assay is used to detect
  • Both of these assays can be used in conjunction with laser scanning cytometry (LSC) to provide both quantitative and morphological analysis of apoptosis.
  • LSC laser scanning cytometry
  • Scanning Cytometry uses lasers to excite fluorochromes in cellular specimens and
  • LSC can additionally find and quantitate events by multiple filter settings, for example, making it possible to distinguish cytoplasmic fluorescence from nuclear
  • LSC has been used to study apoptosis of adipocytes and has been shown to
  • AV Annexin V
  • PI propidium iodide
  • both dyes can be detected at the same time in a single sample.
  • the microscope stage moves the slide (e.g., incubation dish) automatically through
  • the computer For each segment that is scanned, the computer stores information
  • Another method to detect apoptosis is based on the staining of cell suspensions
  • MAbs monoclonal antibodies to single-stranded DNA
  • TUNEL detects low-mol-wt DNA fragmentation associated with late apoptosis
  • MAbs to ssDNA detect the early stages of apoptosis and stain apoptotic cells
  • adipocytes in bone marrow is a major factor contributing to age-related bone loss.
  • mesenchymal stem cells within bone marrow can differentiate to form adipocytes or
  • osteoclastogenic cytokines such as JX-6 (J Clin Endo Metab 83(3): 847-850 (1998), and adipocytes can inhibit osteoblast activity in culture ( " Bone 26(5): 485-9 (2000)).
  • fat cell development and hypertrophy can compress intraosseous capillaries
  • the present disclosure provides compounds, such as, garlic extract
  • Garlic extract compounds of the formula (1) for inducing apoptosis of adiposites.
  • present disclosure can include, but are not limited to, garlic thiosulfinates and transformation products thereof, ajoene (E and Z isomers) (4,5,9-trithiadodeca-l,6,ll- triene-9-oxide), precursors thereof, and derivatives thereof; allicin, precursors thereof,
  • the garlic extract compound can include, but is not limited to, S-
  • SAMC allylmercaptocysteme
  • SMC S-methylcysteine
  • SACS S-allyl cysteine sulfoxide
  • DADS diallyl disulfide
  • SAC S-allylcysteine
  • SEC S-ethylcysteine
  • extract compound includes ajoene (E and Z isomers), and/or precursors thereof, and/or
  • garlic extract compounds may include garlic extract compound analogues, homologues, isomers, or derivatives thereof, that function to
  • garlic extract compounds can include pharmaceutically acceptable salts, esters, and prodrugs of the garlic extract compounds described above.
  • embodiments of this disclosure include methods to treat conditions such as, but not
  • compositions and dosage forms of the disclosure include a
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., oral, mucosal, nasal, sublingual, vaginal, buccal, or rectal
  • dosage forms include,
  • tablets but are not limited to: tablets; caplets; capsules, such as hard gelatin capsules and soft
  • ointments ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters;
  • solutions for solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms
  • suspensions e.g.,
  • aqueous or non-aqueous liquid suspensions oil-in-water emulsions, or water-in-oil
  • liquid emulsions liquid emulsions
  • solutions liquid elixirs
  • liquid dosage forms suitable for parenteral
  • sterile solids e.g., crystalline or amorphous solids
  • compositions of the invention are administered to a patient.
  • shape, and type of dosage forms of the compositions of the invention are administered to a patient.
  • the acute treatment of a condition or disorder may contain larger amounts of the active
  • parenteral dosage form may contain smaller amounts of the active ingredient than an
  • oral dosage form used to treat the same condition or disorder.
  • Typical pharmaceutical compositions and dosage forms include one or more
  • excipients are well known to those skilled in the art of pharmacy
  • composition or dosage form depends on a variety of factors well
  • oral dosage forms such as tablets or
  • capsules may contain excipients not suited for use in parenteral dosage forms.
  • the decomposition of some active ingredients can be any suitable active ingredients.
  • the decomposition of some active ingredients can be any suitable active ingredients.
  • ingredients that include primary or secondary amines are particularly susceptible to
  • the disclosure further encompasses pharmaceutical compositions and dosage
  • antioxidants include, but are not limited to, antioxidants such as ascorbic acid, pH
  • the disclosure may contain one or more solubility modulators, such as sodium
  • exemplary solubility modulator is tartaric acid.
  • the amounts and specific type of active ingredient in a dosage form may differ depending on various factors. It will be understood that the amounts and specific type of active ingredient in a dosage form may differ depending on various factors. It will be understood that the amounts and specific type of active ingredient in a dosage form may differ depending on various factors. It will be understood
  • compositions of the present disclosure are preferably formulated in dosage
  • Dosage unit form for ease of administration and uniformity of dosage.
  • Each dosage should contain the quantity of composition calculated to produce the desired therapeutic affect either as such, or in association with the
  • Preferred unit dosage formulations are those containing a daily dose or unit
  • the approximation includes host factors such as surface area, weight,
  • compositions of the disclosure that are suitable for oral
  • administration can be presented as discrete dosage forms, such as, but not limited to,
  • tablets including without limitation scored or coated tablets
  • pills including without limitation scored or coated tablets
  • caplets including without limitation caplets, capsules
  • chewable tablets powder packets, cachets, troches, wafers, aerosol sprays, or liquids
  • liquid a non-aqueous liquid, an oil-in- water emulsion, or a water-in-oil emulsion).
  • compositions contain a predetermined amount of the pharmaceutically
  • compositions of the disclosure are prepared
  • Excipients can take a wide variety of forms, depending on the form of the composition desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to,
  • tablets can be coated by standard aqueous or nonaqueous techniques. These dosage forms can be prepared by any of the methods of
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing, in a suitable machine, the active
  • ingredient(s) in a free- flowing form such as a powder or granules, optionally mixed
  • Molded tablets can be made by molding, in a suitable
  • Binders include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders
  • compositions and dosage forms suitable for use in pharmaceutical compositions and dosage forms include, but are not
  • gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the
  • suitable binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581. Suitable anhydrous or low moisture excipients or
  • additives include AVICEL-PH- 103TM and Starch 1500 LM.
  • dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate
  • granules or powder e.g., granules or powder
  • microcrystalline cellulose e.g., microcrystalline cellulose, powdered cellulose, dextrates
  • binder or filler in pharmaceutical compositions of the disclosure typically includes
  • Disintegrants may be used in the compositions of the disclosure to provide
  • Tablets that disintegrate when exposed to an aqueous environment Tablets that contain too much disintegrant may swell, crack, or disintegrate in storage, while those
  • the amount of disintegrant used varies, based upon the type
  • Typical pharmaceutical compositions comprise from about 0.5 to 15 weight percent of disintegrant, or from about 1 to 5 weight percent of
  • Disintegrants that can be used to form pharmaceutical compositions and
  • dosage forms of the disclosure include, but are not limited to, agar-agar,, alginic acid,
  • pre-gelatinized starch clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used to form pharmaceutical compositions and dosage
  • forms of the disclosure include, but are not limited to, calcium stearate, magnesium
  • glycol other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable
  • oil e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and
  • soybean oil soybean oil
  • zinc stearate ethyl oleate
  • ethyl laureate ethyl laureate
  • agar agar
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200,
  • lubricants are typically used in an amount of less than about 1 weight
  • compositions preferably contain little, if any, lactose
  • lactose-free means that the amount of lactose present, if any, is insufficient to substantially increase the
  • Lactose-free compositions of the disclosure can comprise excipients that are
  • lactose-free compositions comprise a
  • Preferred lactose-free dosage forms comprise a pharmaceutically acceptable salt of the
  • microcrystalline cellulose microcrystalline cellulose
  • pre-gelatinized starch pre-gelatinized starch
  • magnesium stearate magnesium stearate
  • compositions and dosage forms are provided.
  • lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or
  • An anhydrous pharmaceutical composition should be prepared and stored such
  • anhydrous compositions are provided.
  • compositions of the disclosed compounds can be administered by controlled- or delayed-release means. Controlled-release
  • controlled-release formulations include: 1)
  • Conventional dosage forms generally provide rapid or immediate drug release
  • controlled-release formulations can be used to control a drug's onset
  • ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological conditions
  • hydroxypropylmethyl cellulose examples include hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable
  • multilayer coatings microparticles, liposomes, or microspheres, or a
  • ion exchange materials can be used to prepare immobilized, adsorbed
  • anion exchangers examples include, but are not limited to, Duolite® A568 and Duolite® AP 143 (Rohm & Haas, Spring House, Pa. USA).
  • a pharmaceutically acceptable salt of the disclosed compounds e.g., a
  • form is formulated for controlled-release.
  • Specific dosage forms utilize an osmotic valve
  • a particular and well-known osmotic drug delivery system is referred to as
  • OROS® that can be used to administer compounds and compositions of the disclosure include, but are not limited to, the OROS® Push- PullTM, Delayed Push-PullTM, Multi-Layer Push-PullTM, and Push-StickTM Systems, all
  • compositions of the disclosure include, but are not limited to, OROS®-CT and L-
  • delivery rate of the drug is not influenced by physiological or experimental conditions.
  • a specific dosage form of the compositions of the disclosure includes at least
  • a wall defining a cavity, the wall having an exit orifice formed or
  • a dry or substantially dry state drug layer located within the cavity adjacent the exit orifice and in direct or indirect contacting relationship with the expandable layer; and a flow-promoting layer
  • drug layer located within the cavity, wherein the drug layer includes the compound of
  • Another specific dosage form of the disclosure includes at least the following: a wall defining a cavity, the wall having an exit orifice formed or formable therein and
  • the drug layer comprising a liquid, active-agent formulation absorbed in porous
  • active agent formulation includes the compound of the disclosure, a salt thereof, or a
  • the dosage form also optionally has a placebo layer between the exit orifice
  • Parenteral dosage forms can be administered to patients by various routes,
  • parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient.
  • parenteral dosage forms include, but are not limited to, solutions
  • controlled-release parenteral dosage forms can be prepared for
  • administration to a patient including, but not limited to, administration DUROS®-
  • sterile water water for injection USP; saline solution; glucose solution;
  • aqueous vehicles such as, but not limited to, sodium chloride injection, Ringer's
  • polyethylene glycol, and propylene glycol polyethylene glycol, and propylene glycol
  • non-aqueous vehicles such as, but not
  • Topical dosage forms of the disclosure include, but are not limited to, creams,
  • lotions, ointments, gels, shampoos, sprays, aerosols, solutions, emulsions, and other forms know to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences,
  • topical dosage forms viscous to semi-solid or solid forms comprising a carrier or one
  • excipients compatible with topical application and having a dynamic viscosity
  • Suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders,
  • dosage forms include sprayable aerosol preparations wherein the active ingredient,
  • a pressurized volatile e.g., a gaseous propellant, such as Frean®
  • a pressurized volatile e.g., a gaseous propellant, such as Frean®
  • Fragrance and/or other cosmetic ingredients such as
  • tint, light reflectors, firming agents, and the like can also be added to topical dosage
  • ophthalmic solutions include, but are not limited to, ophthalmic solutions, patches, sprays, aerosols, creams,
  • lotions for example, lotions, suppositories, ointments, gels, solutions, emulsions, suspensions, or other
  • Dosage forms suitable for treating mucosal tissues within the oral cavity can be
  • dosage forms include "reservoir type” or “matrix type” patches, which can be applied'
  • transdermal dosage forms and methods of administration that can be used to produce transdermal dosage forms and methods of administration.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be
  • isopropyl myristate isopropyl palmitate, mineral oil, and mixtures thereof.
  • enhancers can be used to assist in delivering the active ingredients to or across the
  • Suitable penetration enhancers include, but are not limited to: acetone; various others.
  • alcohols such as ethanol, oleyl, an tetrahydrofuryl; alkyl sulfoxides such as dimethyl
  • pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone);
  • TWEEN 80 polysorbate 80
  • SPAN 60 sorbitan monostearate
  • pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be
  • solvent carrier its ionic strength, or tonicity can be adjusted to improve delivery.
  • compositions can also be added to pharmaceutical compositions or
  • stearates can serve as a lipid
  • the active ingredient(s) of the pharmaceutical compositions of the invention preferably, the active ingredient(s) of the pharmaceutical compositions of the
  • disclosure are preferably not administered to a patient at the same time or by the same
  • kits which, when used
  • a typical kit includes a unit dosage form of a pharmaceutically acceptable salt
  • kits may further include ⁇ a device
  • syringes include, but are not limited to, syringes, drip bags, patches, and inhalers.
  • Kits of the disclosure can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients.
  • pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral
  • the kit can include a sealed container of a suitable vehicle in which the
  • active ingredient can be dissolved to form a particulate-free sterile solution that is
  • vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles
  • water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol,
  • non-aqueous vehicles such as, but not limited to, corn oil,
  • PBS Phosphate-buffered saline
  • DMEM medium purchased from GIBCO (BRL Life Technologies, Grand Island, NY). Ajoene was
  • NAC cysteine
  • polyclonal anti-AIF, caspase-3, and PARP-I were from Santa Cruz
  • ERKl/2 (ThT 202 ZTyT 204 ), and total ERK1/2 were from Cell Signaling Technology
  • DMEM fetal calf serum
  • BCS bovine calf serum
  • All media contained 100 U/ml of penicillin, 100 ⁇ g/ml of streptomycin, and of 292
  • MTS cell viability assay Tests were performed in 96-well plates. For mature
  • adipocytes adipocytes
  • Adipocytes were incubated with either DMSO or increasing concentrations of
  • the absorbance was measured at 490 nm in a plate reader ( ⁇ QuantTM
  • Apoptosis assays For the assessment of apoptosis, we used the ApoStrandTM
  • Kit detects single stranded DNA, which occurs in apoptotic cells but not in necrotic
  • adipocytes were incubated with either DMSO or increasing concentrations of ajoene
  • Caspase-3/7 activity was measured using the substrate DEVD- aminoluciferin from the Caspase-GloTM 3/7 assay kit according to the manufacturer's
  • DCHF Molecular Probes, Eugene, OR
  • a lysis buffer (20 mM Tris, pH 7.5, 150 niM NaCl, 1 niM
  • glycerophosphate 1 mM Na 3 VO 4 , and 100 ug/ml phenylmethylsulfonyl fluoride.
  • Tris/glycerol buffer pH 8.5
  • alkaline-phosphatase-conjugated secondary antibody was added.
  • the target proteins became visible following the addition of 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium (BCIP/NBT), a substrate of alkaline phosphatase.
  • BCIP/NBT 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium
  • PVDF membranes after Western blotting with various antibodies was performed using
  • isotonic homogenization buffer 250 mM sucrose, 10 mM
  • KCl 1.5 mM MgCl 2 , 1 mM Na-ethyleneglycotetraacetic acid [EGTA], 1 mM NaOH
  • nuclei and heavy mitochondria fractions were fractionated at 75Og for 10 minutes and
  • mitochondrial and nuclear fractions was measured by Western blot as described above.
  • Tris/EDTA buffer (10 mM Tris-HCl
  • the number of live cells was determined by MTS assay. As shown in FIG. 1, ajoene time and dose-dependently reduced viability in adipocytes. Ajoene at 200
  • Ajoene triggers apoptosis by oxidative stress: To determine the involvement of
  • ROS in ajoene-induced apoptosis ROS levels were determined in ajoene-treated
  • adipocytes As shown in FIG. 3A, ajoene increased ROS production by 2.1 fold after
  • catalase (a scavenger for hydrogen peroxide) did not prevent the ajoene-induced
  • Ajoene induces activation of MAPKs: Since the apoptotic effects of ajoene
  • ROS ROS
  • FIG. 4A shows that activation of JNK occurred as late as 60 min with maximum
  • FIG. 4B shows the time-dependent phosphorylation
  • Ajoene induces PARP-J cleavage and AIF-mediated cell death in a caspase-
  • AIF apoptosis-inducing factor
  • the present example elucidates the biological effect of ajoene, a component of
  • ERK activation is associated with apoptosis induction, whereas ERK activation is
  • ROS levels peaked 20 min after ajoene treatment, whereas ERKl/2 was activated at 30 min and JNK was activated 180 min, which suggests that within this context, ROS
  • JNK protein expression suggests that ROS directly influences MAPK signaling.
  • Catalase is a scavenger for hydrogen peroxide and did not block ajoene-induced ROS generation.
  • NAC is a thiol-reducing agent in addition to its action as a free
  • radical scavenger (31) Ajoene, which contains thiol groups, may be reduced by NAC and thereby lose its ability to induce apoptosis through generation of ROS and subsequent MAPK activation.
  • treatment is caspase independent.
  • NAC inhibited AIF translocation to the nucleus, suggesting that the increased intracellular ROS level is critical for the AIF relocalization after ajoene treatment.
  • PARP is a nuclear enzyme that facilitates DNA repair in response to DNA
  • PARP poly(ADP-ribose) polymerase
  • ROS Reactive oxygen species
  • intracellular ROS level The enhancement of intracellular ROS level promotes
  • ajoene may be a new therapeutic tool for the treatment of obesity by regulating fat cell number through the induction of adipocyte apoptosis.

Abstract

Methods of treating obesity, methods for inducing apoptosis in adipose tissue cells in a host, pharmaceutical compositions, and methods for treating osteoporosis in a host, are disclosed.

Description

COMPOSITIONS AND METHODS FOR INDUCING ADIPOSE TISSUE CELL DEATH
CROSS REFERFENCE TO RELATED APPLICATION
The present application claims priority to and the benefit of U.S. provisional
patent application No. Serial 60/614,738, filed on September 30, 2004 and entitled
"Compositions and Methods for Inducing Adipose Tissue Cell Death", which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
The present disclosure is generally related to compositions and methods for
administration of the compositions to hosts and, more particularly, is related to
compositions designed for treatment for inducing adipose tissue cell death and
methods of administration thereof.
BACKGROUND
Obesity represents a major public health issue that continues to grow, accounts
for 5.7% of total direct health care costs in the United States, and increases the risk of acquiring a disease that is a leading cause of death {e.g., cardiovascular disease,
diabetes, and cancer). Obesity is marked by excess adipose (i.e., fat) tissue
accumulation, which arises from both an increase in number and size of adipocytes
due to higher levels of lipid storage. Excess adipose tissue is strongly correlated with
numerous health problems, including diabetes (e.g., decreased insulin sensitivity),
vascular disease (e.g., hypertension), and certain forms of cancer. Exacerbating the health risks associated with obesity, the most popular treatment for morbid obesity, liposuction, is a largely unregulated, half-billion dollar
industry that exposes patients to additional health risks, including infection and death.
As the current strategy for adipose reduction, liposuction is an invasive and painful
procedure requiring costly equipment and considerable recovery time that often results
in inconsistent tissue shape. Gastric bypass surgery is another extreme treatment for
obesity that is rapidly gaining popularity. Like liposuction, gastric bypass surgery is fraught with risks. It is an even more invasive and serious procedure that requires
extensive recovery time, has a high risk of complications, including death, both during and after surgery, and is too expensive for most potential candidates. Thus, a less
invasive, safer, less expensive, and more reliable way of treating obesity is needed.
Approximately 10 million people in the U.S. are estimated to have
osteoporosis, a disease that results in over 1.5 million bone fractures a year. The
direct expenditures for osteoporosis in 2001 totaled $17 billion, which equals a cost of
$47 million per day. Osteoporosis is therefore a significant health problem, and
effective ways to treat and prevent osteoporosis are needed.
SUMMARY
Methods of treating obesity, methods for inducing apoptosis in adipose tissue
cells in a host, pharmaceutical compositions, and methods for treating osteoporosis in
a host, are disclosed. An illustrative embodiment of a method of treating obesity,
among others, includes administering to a host in need of treatment an effective
amount of a compound selected from: ajoene, precursors thereof, and derivatives
thereof. The compound is present in a dosage level effective to initiate the release or
activation of an apoptosis inducing factor. The apoptosis inducing factor leads to the
apoptotic cell death of adipose tissue cells in the host. An illustrative embodiment of a method for inducing apoptosis in adipose
tissue cells in a host, among others, includes initiating the release or activation of an
apoptosis inducing factor by administering an effective amount of at least one garlic
extract compound. The apoptosis inducing factor leads to apoptotic cell death.
An illustrative embodiment of a pharmaceutical composition, among others,
includes at least one garlic extract compound in combination with a pharmaceutically
acceptable carrier. The at least one garlic extract compound is present in a dosage
level effective to initiate the release of an apoptosis inducing factor. The apoptosis inducing factor leads to the apoptotic cell death of adipose tissue cells in a host.
An illustrative embodiment of a method for treating osteoporosis in a host,
among others, includes administering an effective amount of at least one garlic extract
compound to a host in need of treatment.
An illustrative embodiment of a method for treating osteoporosis in a host,
among others, includes administering to a host an effective amount of a
pharmaceutical composition including at least one garlic extract compound in combination with a pharmaceutically acceptable carrier. The garlic extract compound
is present in a dosage level effective to treat osteoporosis.
Other compositions, methods, features, and advantages of the present
disclosure will be or become apparent to one with skilled in the art upon examination
of the following drawings and detailed description. It is intended that all such
additional compositions, methods, features, and advantages be included within this
description, be within the scope of the present disclosure, and be protected by the
accompanying claims. BRIEF DESCRIPTION OF THE DRAWINGS
Many aspects of the disclosure can be better understood with reference to the
following drawings.
FIG. 1 illustrates the effect of ajoene on cell viability. 3T3-L1 adipocytes were
incubated with ajoene at various concentrations (0, 50, 100, 200, 400 μM) for 6, 12
and 24 h. Cell viability after ajoene treatment was determined by the MTS
colorimetric assay. Assays were performed in eight replicates for each treatment. Within a time period, means with different letters are different, abc: p<0.05; wxyz:
p<0.01.
FIGS. 2A and 2B illustrate the effect of ajoene on apoptosis. In FIG. 2A 3T3-
Ll adipocytes were treated with ajoene at various concentrations (0, 10, 50, 200, 400
μM) for 3, 6, 12 and 24 h. Cells were fixed and analyzed by ssDNA ELISA. Assays
were performed in eight replicates for each treatment. Within a time period, means
with different letters are different, xy: p<0.01. FIG. 2B illustrates apoptosis by gel
electrophoresis. 3T3-L1 adipocytes were incubated with ajoene at various
concentrations (0, 100, 200, 400 μM) for 24 hr. Total cellular DNA was extracted and
analyzed by gel electrophoresis; Lane 1, control; Lane 2, 100 μM ajoene; Lane 3, 200
μM ajoene; Lane 4, 400 μM ajoene; Lane 5, DNA Marker.
FIG. 3 A and 3B illustrate the effects of ajoene on intracellular hydrogen
peroxide production. FIG. 3 A illustrates the time course of the effect of ajoene on
ROS production. 3T3-L1 adipocytes were grown in 96-well plates and treated with
200 μM ajoene, and the change in fluorescence of the oxidized probe was determined.
Each value is the mean ± SEM of eight replicates. Means with different letters are
different (vwxyz), p<0.01. FIG. 3B illustrates the reduction of ajoene-induced apoptosis in cells pretreated with NAC. The percentage of apoptotic cells measured
as described in FIG. 2A is shown for untreated cells (control); cells exposed to ajoene
(200 μM, 24hr); cells preloaded with 10 mM NAC for Ih, either exposed or not
exposed to ajoene (200 μM, 24hr); and cells preloaded with catalase (400 units/ml) in
the absence or presence of ajoene (200 μM, 24hr). Assays were performed in eight
replicates for each treatment. Means with different letters are different (x,y), p<0.01.
FIGS. 4A-4D illustrate the effect of ajoene on MAPKs phosphorylation. The protein levels of unphosphorylated and phosphorylated forms of MAP kinases were
evaluated in cytosolic proteins by Western blotting with the use of specific antibodies.
For the experiments shown in FIG. 4A and 4B, 3T3-L1 adipocytes were treated with
ajoene (200 μM) for the indicated time periods. Representative Western blots show
the time-dependent phosphorylation of (FIG. 4A) JNK and (FIG. 4B) ERK1/2 in the
upper panels and the expression levels of the respective total kinases in the lower
panels.
For the experiments shown in FIG. 4C and FIG. 4D, 3T3-L1 adipocytes were
preincubated with 10 mM NAC for 1 hr before ajoene (200 μM) treatment for either
30 min (for ERK1/2) or 3 hours (for JNK). Western blots showed ERK 1/2 and JNK
phosphorylation (upper panels) as well as the respective total kinases (lower panels).
Densitometric quantitation of the autoradiograms for phosphorylated and total
ERKl/2 and JNK were performed. Integrated density values (phosphorylated/total)
were calculated and expressed as % 0 h (A & B) or % of control (C & D). AU
experiments were repeated two to four times. The data were analyzed by ANOVA
followed by post-hoc analysis by LSD tests. Means with different letters are different, abc: p<0.05; xyz: pθ.01. FIGS. 5A-5C illustrate the effect of ajoene on PARP cleavage and caspase3
activation. In FIG. 5A 3T3-L1 adipocytes were treated with ajoene (200 μM) for the
indicated time periods. Cell lysates were analyzed by Western blotting, and PARP
(upper panel) and caspase-3 protein band (middle panel) were detected using specific
antibodies. β-Actin (lower panel) was used as an equal loading control.
Densitometric quantitation of the autoradiograms for PARP and caspase-3 were
performed. Integrated density values (PARP or caspase/actin) were calculated and expressed as % highest value. All experiments were repeated two to-four times. The
data were analyzed by ANOVA followed by posthoc analysis by LSD tests. Means
with different letters are different: abc, p<0.05; xyz, p<0.01.
In FIG. 5B 3T3-L1 adipocytes were preincubated with 10 mM NAC for 1 hr
before ajoene treatment (200 μM, 12hr). Cell lysates were analyzed by Western
blotting and PARP protein band (upper panel) was detected using specific antibodies.
β-Actin (lower panel) was used as an equal loading control. Densitometric
quantitation of the autoradiogram of PARP was performed. Integrated density values
(PARP/actin) were calculated and expressed as % highest value. AU experiments
were repeated two to four times. The data were analyzed by ANOVA followed by posthoc analysis by LSD tests. Means with different letters are different, xy: p<0.01.
In FIG. 5C 3T3-L1 adipocytes were treated with ajoene at various
concentrations (0, 50, 100, 200 μM) for 1, 3, 6, 12 and 24 hr. Cells were assayed for
caspase-3 activity. Assays were performed in eight replicates for each treatment.
Within a time period, means with different letters are different, xyz: p<0.01.
FIGS. 6A-6B illustrate ajoene induced translocation of AIF from mitochondria
to nucleus. In FIG. 6A 3T3-L1 adipocytes were treated with ajoene (200 μM) for 0, 1, 4, 8 and 12 hr. Equal amounts of AIF protein from nuclear fraction (upper panel) and
mitochondrial fraction (lower panel) were analyzed by Western blotting using an anti-
AIF antibody. Densitometric quantitation of the autoradiogram was performed.
Integrated density values (AIF/actin) were calculated and expressed as % highest
value. Experiments were repeated two times. The data were analyzed by ANOVA
followed by posthoc analysis by LSD tests. Within an AIF source (nuclear or
mitochondrial) means with different letters are different across time: xyz: p<0.01.
In FIG. 6B 3T3-L1 adipocytes were preincubated with 10 mM NAC for 1 hr in
the absence or presence of ajoene (200 μM, 8 hr). Cell lysates were analyzed by
Western blotting using the ami- AIF antibody (upper panel) and β-Actin (lower panel),
which served as an internal control. Densitometric quantitation of the autoradiogram
was performed. Integrated density values (AIF/actin) were calculated and expressed
as % control. Experiments were repeated two times. The data were analyzed by
ANOVA followed by posthoc analysis by LSD tests. Means with different letters are different: xy: p<0.01.
DETAILED DESCRIPTION
Embodiments of the present disclosure will employ, unless otherwise indicated,
techniques of synthetic organic chemistry, biochemistry, molecular biology, and the like,
that are within the skill of the art. Such techniques are explained fully in the literature.
The following examples are put forth so as to provide those of ordinary skill in
the art with a complete disclosure and description of how to perform the methods and
use the compositions disclosed and claimed herein. Efforts have been made to ensure
accuracy with respect to numbers (e.g., amounts, temperature, etc.) but some errors
and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in °C, and pressure is at or near atmospheric. Standard
temperature and pressure are defined as 20 °C and 1 atmosphere.
Before the embodiments of the present disclosure are described in detail, it is
to be understood that, unless otherwise indicated, the present disclosure is not limited
to particular materials, reagents, reaction materials, manufacturing processes, or the
like, as such can vary. It is also to be understood that the terminology used herein is
for purposes of describing particular embodiments only, and is not intended to be limiting. It is also possible in the present disclosure that steps can be executed in
different sequence where this is logically possible.
It must be noted that, as used in the specification and the appended claims, the
singular forms "a," "an" and "the" include plural referents unless the context clearly
dictates otherwise. Thus, for example, reference to "a support" includes a plurality of
supports.
Definitions:
hi this specification and in the claims that follow, reference will be made to a
number of terms that shall be defined to have the following meanings unless a contrary intention is apparent.
The term "organism" or "host" refers to any living entity comprised of at least
one cell. A living organism can be as simple as, for example, a single eukaryotic cell
or as complex as a mammal, including a human being. As used herein, the term
"host" includes humans, mammals (e.g., cats, dogs, horses, chicken, pigs, hogs, cows,
and other cattle), and other living species that are in need of treatment, hi particular,
the term "host" includes humans, companion animals (e.g., cats, dogs, and the like),
and livestock animals (e.g., pigs, cattle, and the like). The term "apoptosis inducing factor" as used herein refers to any compound
activated in a pathway leading to apoptosis. For instance, "apoptosis inducing factor"
includes not only the Apoptosis Inducing Factor protein (AIF), but also any compound
upstream in the pathway leading to ADF release and/or activation, as well as other
compounds which, when activated in certain amounts or under certain conditions lead
to apoptosis. Exemplary apoptosis inducing factors in the present disclosure include,
but are not limited to, reactive oxygen species (ROS), mitogen-activated protein kinases (MAPKs) such as ERK1/2 and INK, PoIy(ADP ribose) polymerases (PARPs),
and AIF. However, apoptosis inducing factors as used in the present disclosure do not
include caspases.
The term "derivative" refers to a modification to the disclosed compounds.
The term "therapeutically effective amount" as used herein refers to that
amount of the compound being administered that will relieve to some extent one or
more of the symptoms of the disorder being treated, hi reference to adipose tissue, a
therapeutically effective amount refers to that amount that has the effect of (1) causing
apoptosis of adipose cells and/or (2) reducing the mass of the adipose cells/tissue.
"Pharmaceutically acceptable salt" refers to those salts that retain the
biological effectiveness and properties of the corresponding free bases and that are
obtained by reaction with inorganic or organic acids such as hydrochloric acid,
hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid,
ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, malic acid, maleic acid,
succinic acid, tartaric acid, citric acid, and the like.
The term "pharmaceutically acceptable esters" as used herein refers to those
esters of one or more compounds of the composition that, within the scope of sound
medical judgment, are suitable for use in contact with the tissues of hosts without undue toxicity, irritation, allergic response, and the like, are commensurate with a
reasonable benefit/risk ratio, and are effective for their intended use.
A "pharmaceutical composition" refers to a mixture of one or more of the
compounds described herein, derivatives thereof, or pharmaceutically acceptable salts
thereof, with other chemical components, such as pharmaceutically acceptable carriers
and excipients. One purpose of a pharmaceutical composition is to facilitate
administration of a compound to the organism.
As used herein, a "pharmaceutically acceptable carrier" refers to a carrier or diluent that does not cause significant irritation to an organism and does not abrogate
the biological activity and properties of the administered compound.
An "excipient" refers to an inert substance added to a pharmaceutical
composition to further facilitate administration of a compound. Examples of
excipients include, without limitation, calcium carbonate, calcium phosphate, various
sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and
polyethylene glycols.
"Treating" or "treatment" of a condition includes preventing the condition
from occurring in an animal that may be predisposed to the disease but does not yet experience or exhibit symptoms of the condition (prophylactic treatment), inhibiting
the condition (slowing or arresting its development), providing relief from the
symptoms or side-effects of the condition (including palliative treatment), and
relieving the condition (causing regression of the condition).
The term "prodrug" refers to an agent that is converted into a biologically
active form in vivo. Prodrugs are often useful because, in some situations, they may
be easier to administer than the parent compound. They may, for instance, be
bioavailable by oral administration whereas the parent compound is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent
drug. A prodrug may be converted into the parent drug by various mechanisms,
including enzymatic processes and metabolic hydrolysis. Harper, NJ. (1962). Drug
Latentiation in Jucker, ed. Progress in Drug Research, 4:221-294; Morozowich et al.
(1977). Application of Physical Organic Principles to Prodrug Design in E. B. Roche
ed. Design of Biopharmaceutical Properties through Prodrugs and Analogs, APhA;
Acad. Pharm. ScL; E. B. Roche, ed. (1977). Bioreversible Carriers in Drug in Drug Design, Theory and Application, APhA; H. Bundgaard, ed. (1985) Design of Prodrugs, Elsevier; Wang et al. (1999) Prodrug approaches to the improved delivery
of peptide drug, Curr. Pharm. Design. 5(4):265-287; Pauletti et al. (1997).
Improvement in peptide bioavailability: Peptidomimetics and Prodrug Strategies, Adv.
Drug. Delivery Rev. 27:235-256; Mizen et al. (1998). The Use of Esters as Prodrugs
for Oral Delivery of β-Lactam antibiotics, Pharm. Biotech. ll,:345-365; Gaignault et
al. (1996). Designing Prodrugs and Bioprecursors I. Carrier Prodrugs, Pract. Med.
Chem. 671-696; M. Asgharnejad (2000). Improving Oral Drug Transport Via
Prodrugs, in G. L. Amidon, P. I. Lee and E. M. Topp, Eds., Transport Processes in Pharmaceutical Systems, Marcell Dekker, p. 185-218; Balant et al. (1990) Prodrugs
for the improvement of drug absorption via different routes of administration, Eur. J.
DrugMetab. Pharmacokinet., 15(2): 143-53; Balimane and Sinko (1999).
Involvement of multiple transporters in the oral absorption of nucleoside analogues,
Adv. Drug Delivery Rev., 39(l-3):183-209; Browne (1997). Fosphenytoin (Cerebyx),
CHn. Neuropharmacol. 20(1): 1-12; Bundgaard (1979). Bioreversible derivatization
of drugs— principle and applicability to improve the therapeutic effects of drugs, Arch.
Pharm. Chemi. 86(1): 1-39; H. Bundgaard, ed. (1985), Design of Prodrugs, New
York: Elsevier; Fleisher et al. (1996). Improved oral drug delivery: solubility limitations overcome by the use of prodrugs, Adv. Drug Delivery Rev. 19(2): 115-130; Fleisher et al. (1985). Design of prodrugs for improved gastrointestinal absorption by
intestinal enzyme targeting, Methods Enzymol. 112: 360-81; Farquhar D, et al. (1983).
Biologically Reversible Phosphate-Protective Groups, J. Pharm. ScL, 72(3): 324-325;
Han, H.K. et al. (2000). Targeted prodrug design to optimize drug delivery, AAPS
PharmScL, 2(1): E6; Sadzuka Y. (2000). Effective prodrug liposome and conversion to active metabolite, Curr. DrugMetab., l(l):31-48; D.M. Lambert (2000). Rationale
and applications of lipids as prodrug carriers, Eur. J. Pharm. ScL, 11 Suppl 2:S15-27; Wang, W. et al. (1999) Prodrug approaches to the improved delivery of peptide drugs.
Curr. Pharm. Des., 5(4):265-87.
As used herein, the term "topically active agents" refers to compositions of the
present disclosure that elicit pharmacological responses at the site of application
(contact) to a host.
As used herein, the term "topically" refers to application of the compositions
of the present disclosure to the surface of the skin and mucosal cells and tissues.
The disclosed compounds can form salts that are also within the scope of this
disclosure. Reference to each compound herein is understood to include reference to
salts thereof, unless otherwise indicated. The term "salt(s)," as employed herein,
denotes acidic and/or basic salts formed with inorganic and/or organic acids and
bases. In addition, when a compound contains both a basic moiety and an acidic
moiety, zwitterions ("inner salts") may be formed and are included within the term
"salt(s)" as used herein. Pharmaceutically acceptable {i.e., non-toxic, physiologically
acceptable) salts are preferred, although other salts are also useful {e.g., in isolation or
purification steps which may be employed during preparation). Salts of the
compounds may be formed, for example, by reacting the compound with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt
precipitates or in an aqueous medium followed by lyophilization.
The disclosed compounds that contain a basic moiety may form salts with a
variety of organic and inorganic acids. Exemplary acid addition salts include acetates
(such as those formed with acetic acid or trihaloacetic acid, for example,
trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates,
benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates,
heptanoates, hexanoates, hydrochlorides (formed with hydrochloric acid),
hydrobromides (formed with hydrogen bromide), hydroiodides, 2-
hydroxyethanesulfonates, lactates, maleates (formed with maleic acid),
methanesulfonates (formed with methanesulfonic acid), 2-naphthalenesulfonates,
nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates (such as
those formed with sulfuric acid), sulfonates (such as those mentioned herein),
tartrates, thiocyanates, toluenesulfonates (such as tosylates), undecanoates, and the
like.
The disclosed compounds that contain an acidic moiety may form salts with a
variety of organic and inorganic bases. Exemplary basic salts include ammonium
salts; alkali metal salts such as sodium, lithium, and potassium salts; alkaline earth
metal salts such as calcium and magnesium salts; salts with organic bases (for
example, organic amines) such as benzathines, dicyclohexylamines, hydrabamines
(formed with N,N-bis(dihydroabietyl)ethylenediamine), N-methyl-D-glucarnines, N- methyl-D-glucamides, t-butyl amines; and salts with amino acids such as arginine,
lysine, and the like.
Basic nitrogen-containing groups may be quaternized with agents such as
lower alkyl halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides and
iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates), long
chain halides (e.g., decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides),
aralkyl halides (e.g., benzyl and phenethyl bromides), and others.
Solvates of the compounds are also contemplated herein. Solvates of the compounds are preferably hydrates.
To the extent that the disclosed compounds, and salts thereof, may exist in
their tautomeric form, all such tautomeric forms are contemplated herein as part of the
present disclosure.
All stereoisomers of the present compounds, such as those which may exist
due to asymmetric carbons on the various substituents, including enantiomeric forms
(which may exist even in the absence of asymmetric carbons) and diastereomeric
forms, are contemplated within the scope of this disclosure. Individual stereoisomers of the compounds of the disclosure may, for example, be substantially free of other
isomers, or may be admixed, for example, as racemates or with all other, or other
selected, stereoisomers. The chiral centers of the compounds of the present disclosure
can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
Discussion
Embodiments of the present disclosure provide for pharmaceutical
compositions, methods for inducing adipose tissue cell death, and methods for treating
obesity. In particular, the compositions include, but are not limited to, compositions having at least one garlic extract compound and, in particular, garlic thiosulfinates and
transformation products thereof. In addition, the method includes inducing adipose
tissue cell death in a host by administering a composition having at least one garlic
extract compound. Further, the methods include treating conditions such as, but not
limited to, obesity and/or osteoporosis, in a host with compositions having at least one
garlic extract compound.
Obesity is a chronic and costly condition that is increasing rapidly throughout the world. Obesity is considered a major risk factor for noninsulin-dependent diabetes
mellitus (1) and has also been linked to cancer and immune dysfunction (2). Adipose tissue mass is determined by processes governing adipocyte size and number (3).
Reduction of adipocyte number can result from preadipocyte and adipocyte apoptosis,
as well as adipocyte de-differentiation (4). Therefore, apoptosis may be an important
mechanism regulating adipose tissue mass.
The garlic extract compound of the present disclosure (e.g., ajoene)
unexpectantly and surprisingly initiates the release of an apoptosis inducing factor,
where the apoptosis inducing factor leads to the apoptotic cell death of adipose tissue cells. This apoptotic cell death is unexpectedly caspase-independent, which is in
contrast to the general understanding that caspases are the primary mediators of
apoptosis under most circumstances. The caspase-independent apoptotic pathway
induced by the compounds of the present disclosure is discussed in more detail in the
Example.
In general, apoptosis refers to a physiological process wherein selected cells
are deleted in a rapid, efficient fashion through a signal-induced activation of
endogenous self-destructive cellular processes. Apoptosis involves a sequence of
distinct biochemical and morphological events characterized by DNA fragmentation, cell volume shrinkage, and production of plasma membrane-bounded apoptosis
bodies, ultimately leading to cell death.
Several organosulfur compounds from garlic, including allicin and its
derivatives, have inhibited the proliferation of some tumor cell lines (5-7). Ajoene is
a garlic-derived compound and has a greater chemical stability than allicin. Ajoene
was shown to induce apoptosis in human promyeloleukemic cells via the activation of
nuclear factor KB (8).
ROS are recognized to play a key role in cell signaling. At the cellular level,
oxidant injury elicits a broad spectrum of responses ranging from proliferation to growth arrest, to senescence, to cell death (for review see 9). Activation of mitogen-
activated protein kinases (MAPK) is considered to be a pivotal step in ROS-induced
signaling pathways. It has been shown that increased ROS production in leukemic
cells leads to the activation of MAPK and cell death (10-13). The MAPK pathways
include three parallel kinase modules, namely, the extracellular signal-regulating
kinase (ERKl/2), the Jun-N-teraiinal kinase (JNK), and the p38 MAPK pathways. In
general, JNK and p38 MAPK activation is associated with apoptosis induction, whereas ERK activation is cytoprotective (14).
Single-strand breaks in DNA, resulting from free radical and oxidant cell
injury, can trigger the activation of the nuclear enzyme, PoIy(ADP ribose) polymerase
(PARP), which contributes to the pathogenesis of various diseases (15, 16). PARP is
a highly conserved, 113 kDa nuclear enzyme which, when activated, generates a
cleaved 85 kDa PARP product.
Apoptosis Inducing Factor (AIF) was more recently cloned and identified as a
mitochondrial intermembrane space protein. In response to apoptotic stimuli, ATF is
released from mitochondria and translocated to the nucleus where it participates in the induction of chromatin condensation, the exposure of phosphatidyl-serine in the outer
leaf of the plasma membrane, and the dissipation of the mitochondrial transmembrane
potential, ultimately resulting in apoptosis (17). Recently, the importance of AIF in
oxidant-induced cell injury in neurons was demonstrated (18). The apoptotic pathway
in the response of 3T3-L1 adipocytes to agents such as ajoene has not previously been
explored, nor have the relationships between components of this pathway.
hi particular, the induction of apoptosis of adipose tissue cells according to the
methods and compounds of the present disclosure is unexpected in that it occurs via the release of apoptosis inducing factors as opposed to the release of caspases. As discussed in more detail in the Example, ajoene-induced apoptosis in 3T3-L1
adipocytes is initiated by the generation of hydrogen peroxide, which leads to
activation of MAPKs, degradation of PARP-I, translocation of AIF, fragmentation of
DNA, and adipose tissue cell death. Therefore, ajoene, a garlic extract compound, can
influence the regulation of fat cell number in a host through the induction of apoptosis
in adipose tissue cells.
As described in more detail in the Example, mature 3T3-L1 adipocytes were
incubated with ajoene at concentrations up to 200 μM. Viability and apoptosis were
quantified using MTS and an ELISA assay for single stranded DNA (ssDNA),
respectively. Intracellular reactive oxygen species (ROS) production was measured
based on production of the fluorescent dye, DCF. Activation of the mitogen-activated
protein kinases (MAPK), extracellular signal-regulating kinase 1/2 (ERK), and c-Jun-
N-terminal kinase (JNK) was demonstrated by Western blot. Western blot was also used to demonstrate activation of caspase-3, translocation of apoptosis inducing factor
(ATF) from mitochondria to nucleus, and cleavage of 116 kDa poly(ADP-ribose)
polymerase (PARP)-I. Ajoene induced apoptosis of 3T3-L1 adipocytes in a dose-and time-dependent
manner. Ajoene treatment resulted in activation of JNK and ERK, translocation of
AIF from mitochondria to nucleus, and cleavage of 116 kDa PARP-I in a caspase-
independent manner. Ajoene treatment also induced an increase in intracellular
reactive oxygen species (ROS) level but did not result in activation of caspases.
Further, the antioxidant N-acetyl-L-cysteine (NAC) effectively blocked ajoene-
mediated'ROS generation, activation of JNK and ERK, translocation of AIF, and degradation of PARP-I. Additional details are described in the Example.
To detect apoptosis, several techniques have been developed based on the understanding of the morphological, biochemical and molecular mechanisms
involved. Changes in the phospholipid bilayers of cell membranes are observed early
in the apoptosis process. The phosphatidylserine (PS) component of the phospholipid
bilayer is externalized and can be detected by fluorescence labeling. Annexin V (AV),
a member of the annexin family of calcium-dependent phospholipid-binding proteins,
has a high affinity for PS-containing phospholipid bilayers. FITC-conjugated AV is
used as a fluorescent dye to detect this early event in apoptotic cells.
As the apoptotic process progresses, cell membranes lose integrity, allowing
chromosomal DNA to be exposed. Propidium iodide, a fluorescent dye that binds to
DNA can be used in conjunction with FITC-conjugated AV to identify subpopulations
of cells with end-stage apoptotic changes.
The TUNEL enzymatic labeling assay is another method used to detect
apoptosis in individual cells. Extensive DNA fragmentation/degradation is a
characteristic event that occurs in apoptosis. The TUNEL assay is used to detect
DNA strand breaks by labeling the free 3'-OH ends. Both of these assays can be used in conjunction with laser scanning cytometry (LSC) to provide both quantitative and morphological analysis of apoptosis. Laser
Scanning Cytometry uses lasers to excite fluorochromes in cellular specimens and
detects the fluorescence in discrete wavelengths with multiple photomultiplier tubes
(PMT's). Data are collected on heterogeneous populations of cells, and software
analysis tools are used to obtain statistical analysis of the populations. LSC also
creates temporary digital images of the specimens on microscope slides and employs image processing algorithms to identify and segment the "events" (e.g., individual
cells). LSC can additionally find and quantitate events by multiple filter settings, for example, making it possible to distinguish cytoplasmic fluorescence from nuclear
fluorescence. Finally, LSC generates high-resolution images that allow visual
inspection of individual cells of interest.
LSC has been used to study apoptosis of adipocytes and has been shown to
provide a relatively fast method for obtaining both quantitative and morphological
information about the apoptotic process in adipocytes.
The two dyes used, Annexin V (AV) and propidium iodide (PI), are
fluorescent dyes that have different absorption spectra, so by using 2 lasers with
different wave lengths, both dyes can be detected at the same time in a single sample. The microscope stage moves the slide (e.g., incubation dish) automatically through
the laser beams. For each segment that is scanned, the computer stores information
about the intensity of the fluorescence from each dye. It also stores digital images of
each segment, which allows one to go back and pick out a point on the scattergraph
and look to see the cells that generated a specific data point.
Another method to detect apoptosis is based on the staining of cell suspensions
and tissue sections with monoclonal antibodies (MAbs) to single-stranded DNA (ssDNA) (Methods MoI Biol 282: 85-102 (2004)). The higher sensitivity of MAb
staining compared to TUNEL reflects the different mechanisms of the two techniques.
TUNEL detects low-mol-wt DNA fragmentation associated with late apoptosis,
whereas MAbs to ssDNA detect the early stages of apoptosis and stain apoptotic cells
in the absence of low-mol-wt DNA fragmentation (Methods MoI Biol 282: 85-102
(2004); Anticancer Res 14(5A): 1861-9 (1994); Exp Cell Res 226(2): 387-97 (1996)). These advantages of the MAb method are based on the fact that protease activation is
an early and universal event in apoptosis (Cell 82(3): 349-52 (1995)). Importantly, in contrast with the TUNEL method, MAbs to ssDNA are specific for apoptotic cell
death and do not detect necrotic cells (Methods MoI Biol 282: 85-102 (2004);
Anticancer Res 14(5A): 1861-9 (1994); Exp Cell Res 226(2): 387-97 (1996)).
In regard to osteoporosis, it is now known that the accumulation of fat cells
(adipocytes) in bone marrow is a major factor contributing to age-related bone loss.
Women with osteoporosis have higher numbers of marrow adipocytes than women
with healthy bone (Clin Orthop 80: 147-54 (1971); J Bone Miner Res 12(11): 1772-9 (1997); and Bio gerontology 2(3): 165-71 (2001)), and bone formation rate is inversely
correlated with adipocyte number in bone tissue biopsies from both men and women
(J Clin Pathol 55(9): 693-8 (2002)). Recent in vivo and in vitro studies provide
important insights into why marrow adipogenesis is associated with bone loss. First,
mesenchymal stem cells within bone marrow can differentiate to form adipocytes or
osteoblasts. Conditions favoring adipocyte differentiation will therefore have adverse
effects on bone formation because precursor cells are directed towards the adipocyte
lineage rather than the osteoblast lineage (J Musculoskel Neuron Interact 2: 581-583
(2002); J Clin Invest 113(6): 846-55 (2004)). Second, adipocytes secrete
osteoclastogenic cytokines such as JX-6 (J Clin Endo Metab 83(3): 847-850 (1998), and adipocytes can inhibit osteoblast activity in culture ("Bone 26(5): 485-9 (2000)).
Finally, fat cell development and hypertrophy can compress intraosseous capillaries,
which decreases blood supply within bone (Joint Bone Spine 69(3): 262-9 (2002)).
Thus, removal of adipocytes from bone marrow through induction of apoptosis could
prevent or reverse bone loss associated with osteoporosis.
The present disclosure provides compounds, such as, garlic extract
compounds, for inducing apoptosis of adiposites. Garlic extract compounds of the
present disclosure can include, but are not limited to, garlic thiosulfinates and transformation products thereof, ajoene (E and Z isomers) (4,5,9-trithiadodeca-l,6,ll- triene-9-oxide), precursors thereof, and derivatives thereof; allicin, precursors thereof,
and derivatives thereof; allyl methanethiosulfinate, precursors thereof, and derivatives
thereof; disulfides, precursors thereof, and derivatives thereof; allylsulfides,
precursors thereof, and derivatives thereof; vinyldithiins, precursors thereof, and
derivatives thereof; diallyl trisulfides, precursors thereof, and derivatives thereof; and
mercaptocysteines, precursors thereof, and derivatives thereof; and combinations
thereof, hi addition, the garlic extract compound can include, but is not limited to, S-
allylmercaptocysteme (SAMC), S-methylcysteine (SMC), S-allyl cysteine sulfoxide (SACS), diallyl disulfide (DADS), S-allylcysteine (SAC), S-ethylcysteine (SEC), S-
propylcysteine (SPC), and combinations thereof, hi one embodiment, the garlic
extract compound includes ajoene (E and Z isomers), and/or precursors thereof, and/or
derivatives thereof.
Where such forms exist, garlic extract compounds may include garlic extract compound analogues, homologues, isomers, or derivatives thereof, that function to
induce adipose tissue cell death in a caspase-independent manner. In addition, garlic extract compounds can include pharmaceutically acceptable salts, esters, and prodrugs of the garlic extract compounds described above.
Embodiments of the present disclosure include methods for inducing apoptosis
of adipose cells and/or reducing the mass of adipose cells/tissue in a host, m addition,
embodiments of this disclosure include methods to treat conditions such as, but not
limited to, obesity, osteoporosis and related conditions and diseases in a host.
Pharmaceutical compositions and dosage forms of the disclosure include a
pharmaceutically acceptable salt of the compound and/or a pharmaceutically acceptable polymorph, solvate, hydrate, dehydrate, co-crystal, anhydrous, or
amorphous form thereof. Specific salts of disclosed compounds include, but are not
limited to, sodium, lithium, and potassium salts, and hydrates thereof.
Pharmaceutical unit dosage forms of the compounds of this disclosure are
suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral
(e.g., intramuscular, subcutaneous, intravenous, intra-arterial, or bolus injection),
topical, or transdermal administration to a patient. Examples of dosage forms include,
but are not limited to: tablets; caplets; capsules, such as hard gelatin capsules and soft
elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories;
ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters;
solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms
suitable for oral or mucosal administration to a patient, including suspensions (e.g.,
aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or water-in-oil
liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral
administration to a patient; and sterile solids (e.g., crystalline or amorphous solids)
that can be reconstituted to provide liquid dosage forms suitable for parenteral
administration to a patient. The composition, shape, and type of dosage forms of the compositions of the
disclosure typically vary depending on their use. For example, a dosage form used in
the acute treatment of a condition or disorder may contain larger amounts of the active
ingredient, e.g., the disclosed compounds or combinations thereof, than a dosage form used in the chronic treatment of the same condition or disorder. Similarly, a
parenteral dosage form may contain smaller amounts of the active ingredient than an
oral dosage form used to treat the same condition or disorder. These and other ways
in which specific dosage forms encompassed by this disclosure vary from one another will be readily apparent to those skilled in the art (See, e.g., Remington's
Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton, Pa. (1990)).
Typical pharmaceutical compositions and dosage forms include one or more
excipients. Suitable excipients are well known to those skilled in the art of pharmacy
or pharmaceutics, and non-limiting examples of suitable excipients are provided
herein. Whether a particular excipient is suitable for incorporation into a
pharmaceutical composition or dosage form depends on a variety of factors well
known in the art including, but not limited to, the way in which the dosage form will
be administered to a patient. For example, oral dosage forms, such as tablets or
capsules, may contain excipients not suited for use in parenteral dosage forms. The
suitability of a particular excipient may also depend on the specific active ingredients
in the dosage form. For example, the decomposition of some active ingredients can
be accelerated by some excipients, such as lactose, or by exposure to water. Active
ingredients that include primary or secondary amines are particularly susceptible to
such accelerated decomposition.
The disclosure further encompasses pharmaceutical compositions and dosage
forms that include one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as
"stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH
buffers, or salt buffers. In addition, pharmaceutical compositions or dosage forms of
the disclosure may contain one or more solubility modulators, such as sodium
chloride, sodium sulfate, sodium or potassium phosphate, or organic acids. An
exemplary solubility modulator is tartaric acid.
Like the amounts and types of excipients, the amounts and specific type of active ingredient in a dosage form may differ depending on various factors. It will be
understood, however, that the total daily usage of the compositions of the present
disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular
host will depend upon a variety of factors, including for example, the disorder being
treated and the severity of the disorder; the activity of the specific composition
employed; the specific composition employed; the age, body weight, general health,
sex, and diet of the host; the time of administration; the route of administration; the
rate of excretion of the specific compound employed; the duration of the treatment;
the existence of other drugs used in combination or coincidental with the specific composition employed; and like factors well known in the medical arts. For example,
it is well within the skill of the art to start doses of the composition at levels lower
than those required to achieve the desired therapeutic effect and to gradually increase
the dosage until the desired effect is achieved.
Compositions of the present disclosure are preferably formulated in dosage
unit form for ease of administration and uniformity of dosage. "Dosage unit form" as
used herein refers to a physically discrete unit of the composition appropriate for the
host to be treated. Each dosage should contain the quantity of composition calculated to produce the desired therapeutic affect either as such, or in association with the
selected pharmaceutical carrier medium.
Preferred unit dosage formulations are those containing a daily dose or unit,
daily sub-dose, or an appropriate fraction thereof, of the administered ingredient. For
example, approximately 8 milligrams/kilogram per day of a garlic extract compound
(e.g., allicin) can reduce the mass of adipose tissue in a rat. These results can be used
to predict an approximate amount of the garlic extract compound to be administered to a human or other host, such as cattle.
The approximation includes host factors such as surface area, weight,
metabolism, tissue distribution, absorption rate, and excretion rate, for example.
Therefore, approximately 1 to 100 milligrams/kilogram per day of the ajoene should
produce similar results in humans. As stated above, a therapeutically effective dose
level will depend on many factors, as described above. In addition, it is well within
the skill of the art to start doses of the composition at relatively low levels, and
increase the dosage until the desired effect is achieved.
Pharmaceutical compositions of the disclosure that are suitable for oral
administration can be presented as discrete dosage forms, such as, but not limited to,
tablets (including without limitation scored or coated tablets), pills, caplets, capsules,
chewable tablets, powder packets, cachets, troches, wafers, aerosol sprays, or liquids,
(such as but not limited to, syrups, elixirs, solutions or suspensions in an aqueous
liquid, a non-aqueous liquid, an oil-in- water emulsion, or a water-in-oil emulsion).
Such compositions contain a predetermined amount of the pharmaceutically
acceptable salt of the disclosed compounds, and may be prepared by methods of
pharmacy well known to those skilled in the art. See generally, Remington's
Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton, Pa. (1990). Typical oral dosage forms of the compositions of the disclosure are prepared
by combining the pharmaceutically acceptable salt of disclosed compounds in an
intimate admixture with at least one excipient according to conventional
pharmaceutical compounding techniques. Excipients can take a wide variety of forms, depending on the form of the composition desired for administration. For
example, excipients suitable for use in oral liquid or aerosol dosage forms include, but
are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches,
sugars, microcrystalline cellulose, kaolin, diluents, granulating agents, lubricants,
binders, and disintegrating agents.
Due to their ease of administration, tablets and capsules represent the most
advantageous solid oral dosage unit forms, in which case solid pharmaceutical
excipients are used. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. These dosage forms can be prepared by any of the methods of
pharmacy, hi general, pharmaceutical compositions and dosage forms are prepared by
uniformly and intimately admixing the active ingredient(s) with liquid carriers, finely
divided solid carriers, or both, and then shaping the product into the desired
presentation if necessary.
For example, a tablet can be prepared by compression or molding.
Compressed tablets can be prepared by compressing, in a suitable machine, the active
ingredient(s) in a free- flowing form, such as a powder or granules, optionally mixed
with one or more excipients. Molded tablets can be made by molding, in a suitable
machine, a mixture of the powdered compound moistened with an inert liquid diluent. Examples of excipients that can be used in oral dosage forms of the disclosure
include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders
suitable for use in pharmaceutical compositions and dosage forms include, but are not
limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic
gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose
acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
Suitable forms of microcrystalline cellulose include, but are not limited to, the
materials sold as AVICEL-PHr101, AVICEL-PH- 103, AVICEL RC-581, and
AVICEL-PH- 105 (available from FMC Corporation, American Viscose Division,
Avicel Sales, Marcus Hook, Pa., U.S.A.), and mixtures thereof. An exemplary
suitable binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581. Suitable anhydrous or low moisture excipients or
additives include AVICEL-PH- 103™ and Starch 1500 LM.
Examples of fillers suitable for use in the pharmaceutical compositions and
dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate
(e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates,
kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures
thereof. The binder or filler in pharmaceutical compositions of the disclosure typically
accounts for from about 50 to 99 weight percent of the pharmaceutical composition or
dosage form.
Disintegrants may be used in the compositions of the disclosure to provide
tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may swell, crack, or disintegrate in storage, while those
that contain too little may be insufficient for disintegration to occur and may thus alter
the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a
sufficient amount of disintegrant that is neither too little nor too much to detrimentally
alter the release of the active ingredient(s) should be used to form solid oral dosage
forms of the disclosure. The amount of disintegrant used varies, based upon the type
of formulation and mode of administration, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to 15 weight percent of disintegrant, or from about 1 to 5 weight percent of
disintegrant.
Disintegrants that can be used to form pharmaceutical compositions and
dosage forms of the disclosure include, but are not limited to, agar-agar,, alginic acid,
calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone,
polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches,
pre-gelatinized starch, clays, other algins, other celluloses, gums, and mixtures thereof.
Lubricants that can be used to form pharmaceutical compositions and dosage
forms of the disclosure include, but are not limited to, calcium stearate, magnesium
stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene
glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable
oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and
soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
Additional lubricants include, for example, a syloid silica gel (AEROSIL 200,
manufactured by W. R. Grace Co. of Baltimore, Md.), a coagulated aerosol of
synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-O-SDL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If
used at all, lubricants are typically used in an amount of less than about 1 weight
percent of the pharmaceutical compositions or dosage forms into which they are
incorporated.
This disclosure further encompasses lactose-free pharmaceutical compositions
and dosage forms, wherein such compositions preferably contain little, if any, lactose
or other mono- or disaccharides. As used herein, the term "lactose-free" means that the amount of lactose present, if any, is insufficient to substantially increase the
degradation rate of an active ingredient.
Lactose-free compositions of the disclosure can comprise excipients that are
well known in the art and are listed in the USP (XXI)/NF (XVI), which is
incorporated herein by reference. In general, lactose-free compositions comprise a
pharmaceutically acceptable salt of a catecholamine compound, a binder/filler, and a
lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
Preferred lactose-free dosage forms comprise a pharmaceutically acceptable salt of the
disclosed compounds, microcrystalline cellulose, pre-gelatinized starch, and
magnesium stearate.
Since water can facilitate the degradation of some compounds, this disclosure
further encompasses anhydrous pharmaceutical compositions and dosage forms of the
disclosed compounds as active ingredients. For example, the addition of water (e.g.,
5%) is widely accepted in the pharmaceutical arts as a means of simulating long-term
storage in order to determine characteristics such as shelf life or the stability of
formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles &
Practice, 379-80 (2nd ed., Marcel Dekker, NY, N. Y.: 1995). Water and heat
accelerate the decomposition of some compounds. Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly
encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
Anhydrous pharmaceutical compositions and dosage forms of the disclosure
can be prepared using anhydrous or low moisture containing ingredients and low-
moisture or low-humidity conditions. Pharmaceutical compositions and dosage forms
that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or
humidity during manufacturing, packaging, and/or storage is expected.
An anhydrous pharmaceutical composition should be prepared and stored such
that its anhydrous nature is maintained. Accordingly, anhydrous compositions are
preferably packaged using materials known to prevent exposure to water such that
they can be included in suitable formulary kits. Examples of suitable packaging
include, but are not limited to, hermetically sealed foils, plastics, unit dose containers
(e.g., vials) with or without desiccants, blister packs, and strip packs.
Pharmaceutically acceptable salts of the disclosed compounds can be administered by controlled- or delayed-release means. Controlled-release
pharmaceutical products have a common goal of improving drug therapy over that
achieved by their non-controlled release counterparts. Ideally, the use of an optimally
designed controlled-release preparation in medical treatment is characterized by a
minimum of drug substance being employed to cure or control the condition in an
appropriate amount of time. Advantages of controlled-release formulations include: 1)
extended activity of the drug; 2) reduced dosage frequency; 3) increased patient
compliance; 4) usage of less total drug; 5) reduction in local or systemic side effects;
6) minimization of drug accumulation; 7) reduction in blood level fluctuations; 8) improvement in efficacy of treatment; 9) reduction of potentiation or loss of drug
activity; and 10) improvement in speed of control of diseases or conditions. See, e.g.,
Kim, Cherng-ju, Controlled Release Dosage Form Design, 2 (Technomic Publishing, Lancaster, Pa.: 2000).
Conventional dosage forms generally provide rapid or immediate drug release
from the formulation. Depending on the pharmacology and pharmacokinetics of the
drug, use of conventional dosage forms can lead to wide fluctuations in the concentrations of the drug in a patient's blood and other tissues. These fluctuations
can impact a number of parameters, such as dose frequency, onset of action, duration of efficacy, maintenance of therapeutic blood levels, toxicity, side effects, and the like.
Advantageously, controlled-release formulations can be used to control a drug's onset
of action, duration of action, plasma levels within the therapeutic window, and peak
blood levels, hi particular, controlled- or extended-release dosage forms or
formulations can be used to ensure that the maximum effectiveness of a drug is
achieved while minimizing potential adverse effects and safety concerns, which can
occur both from under dosing a drug (i.e., going below the minimum therapeutic
levels) as well as from exceeding the toxicity level for the drug.
Most controlled-release formulations are designed to initially release an
amount of drug (active ingredient) that promptly produces the desired therapeutic
effect, and gradually and continually release other amounts of drug to maintain this
level of therapeutic or prophylactic effect over an extended period of time. In order to
maintain a relatively constant level of drug in the body, the drug must be released
from the dosage form at a rate such that newly released drug will replace the amount
of drug being metabolized and excreted from the body. Controlled-release of an active
ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological
conditions or compounds.
A variety of known controlled- or extended-release dosage forms,
formulations, and devices can be adapted for use with the salts and compositions of
the disclosure. Examples include, but are not limited to, those described in U.S. Pat.
Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595;
5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,733,566; and 6,365,185 Bl; each of which is incorporated herein by reference. These dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for
example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable
membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View,
Calif. USA)), multilayer coatings, microparticles, liposomes, or microspheres, or a
combination thereof to provide the desired release profile in varying proportions.
Additionally, ion exchange materials can be used to prepare immobilized, adsorbed
salt forms of the disclosed compounds and thus effect controlled delivery of the drug.
Examples of specific anion exchangers include, but are not limited to, Duolite® A568 and Duolite® AP 143 (Rohm & Haas, Spring House, Pa. USA).
One embodiment of the disclosure encompasses a unit dosage form which
comprises a pharmaceutically acceptable salt of the disclosed compounds (e.g., a
sodium, potassium, or lithium salt), or a polymorph, solvate, hydrate, dehydrate, co-
crystal, anhydrous, or amorphous form thereof, and one or more pharmaceutically
acceptable excipients or diluents, wherein the pharmaceutical composition or dosage
form is formulated for controlled-release. Specific dosage forms utilize an osmotic
drug delivery system. A particular and well-known osmotic drug delivery system is referred to as
OROS® (Alza Corporation, Mountain View, Calif. USA). This technology can
readily be adapted for the delivery of compounds and compositions of the disclosure.
Various aspects of the technology are disclosed in U.S. Pat. Nos. 6,375,978 Bl;
6,368,626 Bl; 6,342,249 Bl; 6,333,050 B2; 6,287,295 Bl; 6,283,953 Bl; 6,270,787
Bl; 6,245,357 Bl; and 6,132,420; each of which is incorporated herein by reference.
Specific adaptations of OROS® that can be used to administer compounds and compositions of the disclosure include, but are not limited to, the OROS® Push- Pull™, Delayed Push-Pull™, Multi-Layer Push-Pull™, and Push-Stick™ Systems, all
of which are well known. See, e.g. worldwide website alza.com. Additional OROS®
systems that can be used for the controlled oral delivery of compounds and
compositions of the disclosure include, but are not limited to, OROS®-CT and L-
OROS® (see, Delivery Times, vol. 11, issue II (Alza Corporation)).
Conventional OROS® oral dosage forms are made by compressing a drug
powder into a hard tablet, coating the tablet with cellulose derivatives to form a semi¬ permeable membrane, and then drilling an orifice in the coating (e.g., with a laser).
Kim, Cherng-ju, Controlled Release Dosage Form Design, 231-238 (Technomic
Publishing, Lancaster, Pa.: 2000). The advantage of such dosage forms is that the
delivery rate of the drug is not influenced by physiological or experimental conditions.
Even a drug with a pH-dependent solubility can be delivered at a constant rate
regardless of the pH of the delivery medium. However, because these advantages are
provided by a build-up of osmotic pressure within the dosage form after
administration, conventional OROS® drug delivery systems cannot be used to
effectively deliver drugs with low water solubility. This disclosure encompass the incorporation of the compounds of the present disclosure and salts thereof, non-salt
isomers and isomeric mixtures thereof and the like into OROS® dosage forms.
A specific dosage form of the compositions of the disclosure includes at least
the following: a wall defining a cavity, the wall having an exit orifice formed or
formable therein and at least a portion of the wall being semipermeable; an
expandable layer located within the cavity remote from the exit orifice and in fluid
communication with the semipermeable portion of the wall; a dry or substantially dry state drug layer located within the cavity adjacent the exit orifice and in direct or indirect contacting relationship with the expandable layer; and a flow-promoting layer
interposed between the inner surface of the wall and at least the external surface of the
drug layer located within the cavity, wherein the drug layer includes the compound of
the disclosure, a salt thereof, or a polymorph, solvate, hydrate, dehydrate, co-crystal,
anhydrous, or amorphous form thereof. See U.S. Pat. No. 6,368,626, the entirety of
which is incorporated herein by reference.
Another specific dosage form of the disclosure includes at least the following: a wall defining a cavity, the wall having an exit orifice formed or formable therein and
at least a portion of the wall being semipermeable; an expandable layer located within
the cavity remote from the exit orifice and in fluid communication with the
semipermeable portion of the wall; a drug layer located within the cavity adjacent the
exit orifice and in direct or indirect contacting relationship with the expandable layer,
the drug layer comprising a liquid, active-agent formulation absorbed in porous
particles adapted to resist compaction forces sufficient to form a compacted drug layer
without significant exudation of the liquid, active-agent formulation, wherein the
active agent formulation includes the compound of the disclosure, a salt thereof, or a
polymorph, solvate, hydrate, dehydrate, co-crystal, anhydrous, or amorphous form thereof. The dosage form also optionally has a placebo layer between the exit orifice
and the drug layer. {See U.S. Pat. No. 6,342,249, the entirety of which is incorporated herein by reference.)
Parenteral dosage forms can be administered to patients by various routes,
including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intra-arterial. Since administration of parenteral dosage forms
typically bypasses the host's natural defenses against contaminants, parenteral dosage
forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions
ready for injection, dry products ready to be dissolved or suspended in a
pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and
emulsions. In addition, controlled-release parenteral dosage forms can be prepared for
administration to a patient, including, but not limited to, administration DUROS®-
type dosage forms, and dose-dumping.
Suitable vehicles that can be used to provide parenteral dosage forms of the
disclosure are well known to those skilled in the art. Examples include, without
limitation: sterile water; water for injection USP; saline solution; glucose solution;
aqueous vehicles such as, but not limited to, sodium chloride injection, Ringer's
injection, dextrose injection, dextrose and sodium chloride injection, and lactated
Ringer's injection; water-miscible vehicles such as, but not limited to, ethyl alcohol,
polyethylene glycol, and propylene glycol; and non-aqueous vehicles such as, but not
limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl
myristate, benzyl benzoate, and mixtures thereof.
Topical dosage forms of the disclosure include, but are not limited to, creams,
lotions, ointments, gels, shampoos, sprays, aerosols, solutions, emulsions, and other forms know to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences,
18th ed., Mack Publishing, Easton, Pa. (1990); and Introduction to Pharmaceutical
Dosage Forms, 4th ed., Lea & Febiger, Philadelphia, Pa. (1985). For non-sprayable
topical dosage forms, viscous to semi-solid or solid forms comprising a carrier or one
or more excipients compatible with topical application and having a dynamic viscosity
preferably greater than water are typically employed. Suitable formulations include, without limitation, solutions, suspensions, emulsions, creams, ointments, powders,
liniments, salves, and the like, which are, if desired, sterilized or mixed with auxiliary agents (e.g., preservatives, stabilizers, wetting agents, buffers, or salts) for influencing
various properties, such as, for example, osmotic pressure. Other suitable topical
dosage forms include sprayable aerosol preparations wherein the active ingredient,
preferably in combination with a solid or liquid inert carrier, is packaged in a mixture
with a pressurized volatile (e.g., a gaseous propellant, such as Frean®), or in a squeeze
bottle. Moisturizers or humectants can also be added to pharmaceutical compositions
and dosage forms if desired. Fragrance and/or other cosmetic ingredients (such as
tint, light reflectors, firming agents, and the like) can also be added to topical dosage
forms, if desired. Examples of such additional ingredients are well known in the art.
See, e.g., Remington's Pharmaceutical Sciences, 18.sup.th Ed., Mack Publishing,
Easton, Pa. (1990).
Transdermal and mucosal dosage forms of the compositions of the disclosure
include, but are not limited to, ophthalmic solutions, patches, sprays, aerosols, creams,
lotions, suppositories, ointments, gels, solutions, emulsions, suspensions, or other
forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical
Sciences, 18th Ed., Mack Publishing, Easton, Pa. (1990); and Introduction to
Pharmaceutical Dosage Forms, 4th Ed., Lea & Febiger, Philadelphia, Pa. (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be
formulated as mouthwashes, as oral gels, or as buccal patches. Additional transdermal
dosage forms include "reservoir type" or "matrix type" patches, which can be applied'
to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredient.
Examples of transdermal dosage forms and methods of administration that can
be used to administer the active ingredient(s) of the disclosure include, but are not limited to, those disclosed in U.S. Pat. Nos.: 4,624,665; 4,655,767; 4,687,481;
4,797,284; 4,810,499; 4,834,978; 4,877,618; 4,880,633; 4,917,895; 4,927,687; 4,956,171; 5,035,894; 5,091,186; 5,163,899; 5,232,702; 5,234,690; 5,273,755;
5,273,756; 5,308,625; 5,356,632; 5,358,715; 5,372,579; 5,421,816; 5,466;465;
5,494,680; 5,505,958; 5,554,381; 5,560,922; 5,585,111; 5,656,285; 5,667,798;
5,698,217; 5,741,511; 5,747,783; 5,770,219; 5,814,599; 5,817,332; 5,833,647;
5,879,322; and 5,906,830, each of which are incorporated herein by reference in their
entirety.
Suitable excipients (e.g., carriers and diluents) and other materials that can be
used to provide transdermal and mucosal dosage forms encompassed by this
disclosure are well known to those skilled in the pharmaceutical arts, and depend on
the particular tissue or organ to which a given pharmaceutical composition or dosage
form will be applied. With that fact in mind, typical excipients used in form dosage
forms that are non-toxic and pharmaceutically acceptable include, but are not limited
to water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1, 3 -diol,
isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof.
Depending on the specific tissue to be treated, additional components may be
used prior to, in conjunction with, or subsequent to treatment with pharmaceutically acceptable salts of the compounds of the disclosure. For example, penetration
enhancers can be used to assist in delivering the active ingredients to or across the
tissue. Suitable penetration enhancers include, but are not limited to: acetone; various
alcohols such as ethanol, oleyl, an tetrahydrofuryl; alkyl sulfoxides such as dimethyl
sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol;
pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone);
urea; and various water-soluble or insoluble sugar esters such as TWEEN 80 (polysorbate 80) and SPAN 60 (sorbitan monostearate).
The pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied, may also be
adjusted to improve delivery of the active ingredient(s). Similarly, the polarity of a
solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
Compounds such as stearates can also be added to pharmaceutical compositions or
dosage forms to advantageously alter the hydrophilicity or lipophilicity of the active
ingredient(s) so as to improve delivery, hi this regard, stearates can serve as a lipid
vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery- enhancing or penetration-enhancing agent. Different hydrates, dehydrates, co-
crystals, solvates, polymorphs, anhydrous, or amorphous forms of the
pharmaceutically acceptable salt of the compounds can be used to further adjust the
properties of the resulting composition.
Typically, the active ingredient(s) of the pharmaceutical compositions of the
disclosure are preferably not administered to a patient at the same time or by the same
route of administration. This disclosure therefore encompasses kits which, when used
by the medical practitioner, can simplify the administration of appropriate amounts of
active ingredients to a patient. A typical kit includes a unit dosage form of a pharmaceutically acceptable salt
of the compound. In particular, the pharmaceutically acceptable salt of the compound
is the sodium, lithium, or potassium salt, or a polymorph, solvate, hydrate, dehydrate,
co-crystal, anhydrous, or amorphous form thereof. A kit may further include^a device
that can be used to administer the active ingredient. Examples of such devices
include, but are not limited to, syringes, drip bags, patches, and inhalers.
Kits of the disclosure can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral
administration, the kit can include a sealed container of a suitable vehicle in which the
active ingredient can be dissolved to form a particulate-free sterile solution that is
suitable for parenteral administration. Examples of pharmaceutically acceptable
vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles
such as, but not limited to, sodium chloride injection, Ringer's injection, dextrose
injection, dextrose and sodium chloride injection, and lactated Ringer's injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol,
and propylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil,
cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl
benzoate.
Having summarized embodiments, reference will now be made in detail to the
illustrative Example. While the disclosure is described in connection with the
Example, there is no intent to limit the embodiments of the disclosure to the following
example. On the contrary, the intent is to cover all alternatives, modifications, and
equivalents included within the spirit and scope of the disclosure. EXAMPLE
This example demonstrates that ajoene-induced ROS generation plays a
central role in the events of the apoptotic pathway and is responsible for activation of
the MAPK pathways. Furthermore, the results indicate that garlic extract compounds such as ajoene play an important role in the translocation of AIF from the
mitochondria to the nucleus, and the results demonstrate the role of PARP in
regulating this process. Materials and Methods:
Chemicals: Phosphate-buffered saline (PBS) and DMEM medium were purchased from GIBCO (BRL Life Technologies, Grand Island, NY). Ajoene was
received as a gift from Rafael Apitz-Castro, Institute for Thrombosis Research,
Caracao, Venezuela. ApoStrand™ ELISA Apoptosis Detection Kit was purchased
from BIOMOL (Plymouth Meeting, PA). The viability assay kit (CellTiter 96
Aqueous One Solution Cell Proliferation Assay) and Caspase-Glo™ 3/7 assay kit
were purchased from Promega (Madison, WI). β-Actin, Catalase and N-acetyl-L-
cysteine (NAC) were purchased from Sigma (St. Louis, MO, USA). Antibodies
specific for polyclonal anti-AIF, caspase-3, and PARP-I were from Santa Cruz
Biotechnology (Santa Cruz, CA). Antibodies specific for polyclonal anti- phospho-
p38 (Thr180/Tyr182), total p38, phospho-JNK (Thr183/Tyr185), total JNK, phospho-
ERKl/2 (ThT202ZTyT204), and total ERK1/2 were from Cell Signaling Technology
(Beverly, MA).
Cell line and Cell culture: 3T3-L1 mouse embryo fibroblasts were obtained
from American Type Culture Collection (Manassas, VA) and cultured as described
elsewhere (20). Briefly, cells were cultured in Dulbecco's modified Eagle's medium
(DMEM) containing 10% bovine calf serum (BCS) until confluent. Two days after postconfluence (DO), the cells were" stimulated to differentiate with DMEM containing
10% fetal bovine serum (FBS), 167 nM insulin, 0.5 μM isobutylmethylxanthine
(IBMX), and 1 μM dexamethasone for two days (D2). Cells were then maintained in
10% FBS/DMEM medium with 167 nM insulin for another two days (D4), followed
by culturing with 10% FBS/DMEM medium for an additional 4 days (DS), at which
time more than 90% of cells were mature adipocytes with accumulated fat droplets.
All media contained 100 U/ml of penicillin, 100 μg/ml of streptomycin, and of 292
μg/ml glutamine (Livitrogen, Carlsbad, CA). Cells were maintained at 37°C in a
humidified 5% CO2 atmosphere.
MTS cell viability assay: Tests were performed in 96-well plates. For mature
adipocytes, cells were seeded (5,000 cells/well) and grown to maturation as described
above. Adipocytes were incubated with either DMSO or increasing concentrations of
ajoene for 6, 12, and 24 h. The medium was then changed and replaced with 100 μl
fresh 10% FBS/DMEM medium and 20 μl MTS solution. Cells were then returned to
the incubator for an additional two hours before 25 μl of 10% SDS was added to stop
the reaction. The absorbance was measured at 490 nm in a plate reader (μQuant™
Bio-Tek Instruments, Inc. Winooski, VT) to determine the formazan concentration,
which is proportional to the number of live cells.
Apoptosis assays: For the assessment of apoptosis, we used the ApoStrand™
ELISA Apoptosis Detection Kit (Biomol, Plymouth Meeting, PA) and Caspase-Glo™
3/7 assay kit (Promega, Madison, WI). The ApoStrand™ ELISA Apoptosis Detection
Kit detects single stranded DNA, which occurs in apoptotic cells but not in necrotic
cells or in cells with DNA breaks in the absence of apoptosis (21, 22). Tests were
performed in 96-well plates. For mature adipocytes, cells were seeded (5,000 cells/well) and grown to maturation as described above. For ssDNA ELISA5
adipocytes were incubated with either DMSO or increasing concentrations of ajoene
for 3, 6, 12 and 24 h. Thereafter, cells were fixed for 30 min and assayed according to
the manufacturer's instructions. For the caspase activity assay, cells were incubated
with either DMSO (carrier; 0.1%) or increasing concentrations of ajoene for 1, 3, 6,
12 and 24 h. Caspase-3/7 activity was measured using the substrate DEVD- aminoluciferin from the Caspase-Glo™ 3/7 assay kit according to the manufacturer's
instructions.
Measurement of intracellular ROS generation: The determination of ROS was
based on the oxidation of the nonfluorescent 2,7-dichlorodihydroflourescein diacetate
(DCHF) into a fluorescent dye, 2,7- dichloroflourescein "(DCF) by peroxide. Control
cells and cells treated with 200 μM ajoene were analyzed for changes in fluorescence.
Cells were washed twice with PBS and then incubated for 30 min at 37 0C in the dark
with the oxidation-sensitive probe, DCHF (Molecular Probes, Eugene, OR) at 2.5 μM.
Production of ROS was measured by changes in fluorescence at an excitation
wavelength of 495 nm and an emission wavelength of 525 ran.
Western blot analysis: To prepare the whole-cell extract, cells were washed
with PBS and suspended in a lysis buffer (20 mM Tris, pH 7.5, 150 niM NaCl, 1 niM
EDTA, ImM EGTA, 1 % Triton X-IOO, 2.5 mM Na pyrophosphate, ImM β-
glycerophosphate, 1 mM Na3VO4, and 100 ug/ml phenylmethylsulfonyl fluoride).
After 30 min of rocking at 4°C, the mixtures were centrifuged (10,00Og) for 10 min,
and the supernatants were collected as the whole-cell extracts. The cytosolic protein
concentration was determined by the method of Bradford (23) with bovine serum
albumin as the standard. Western blot analysis was performed using the commercial NUPAGE system (Novex/Invitrogen), where a lithium dodecyl sulfate (LDS) sample
buffer (Tris/glycerol buffer, pH 8.5) was mixed with fresh dithiothreitol and added to
samples. Samples were then heated to 70°C for 10 min. All cell lysates were
separated by 12% acrylamide gels and transferred to PVDF membranes. The
membranes were blocked with 5% nonfat dry milk in Tris-buffered saline and then
incubated with primary antibodies overnight at 40C. After washing the membranes,
an alkaline-phosphatase-conjugated secondary antibody was added. The target proteins became visible following the addition of 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium (BCIP/NBT), a substrate of alkaline phosphatase.
All experiments were repeated at least two times. Representative Western blots are
shown along with the graphs of the quantitative data.
Quantitative analysis of Western blot data: Measurement of signal intensity on
PVDF membranes after Western blotting with various antibodies was performed using
a FluorChem™densitomer with the AlphaEaseFC™ image processing and analysis
software (Alpha Innotech Corporation). In FIG. 4 density values for the protein bands
of interest are expressed as percentage of the control or 0 h. In FIGS. 5 A and B and 6A, density values are expressed as percent of the highest value in order to show more
clearly how the levels of each protein change. In FIG. 6B density values are expressed
as percent of control. AU figures showing quantitative analysis include data from at
least two independent experiments.
Preparation of nuclear and mitochondrial fractions for measurement of AIF
by Western blot: The cells were washed with ice-cold PBS, left on ice for 10 minutes,
and then resuspended in isotonic homogenization buffer (250 mM sucrose, 10 mM
KCl, 1.5 mM MgCl2, 1 mM Na-ethyleneglycotetraacetic acid [EGTA], 1 mM NaOH
(Na)-ethylenediaminetetraacetic acid [EDTA], 1 mM dithiothreitol, 0.1 mM phenylmethylsulfonylfluoride, 10 mM HEPES-KOH, pH 7.4). After 80 strokes in a
Dounce homogenizer, the unbroken cells were spun down at 30g for 5 minutes. The
nuclei and heavy mitochondria fractions were fractionated at 75Og for 10 minutes and
14,00Og for 20 minutes, respectively, from the supernatant. The nuclei fraction was
washed 3 times with homogenization buffer containing 0.01% NP-40. AIF in
mitochondrial and nuclear fractions was measured by Western blot as described above.
Detection of DNA fragmentation by gel electrophoresis: Cell pellets (3 x 106
cells) were resuspended in 500 μl of lysis buffer (0.5% Triton X-100, 10 mM EDTA,
and 10 mM Tris-HCl, pH 8.0) at room temperature for 15 min and centrifuged at
16,00Og for 10 min. DNA was then extracted twice with phenolxhloroform (1:1),
precipitated with ethanol, and resuspended in Tris/EDTA buffer (10 mM Tris-HCl,
pH 8.0, and 1 mM EDTA). DNA was analyzed after separation by gel electrophoresis
(2% agarose).
Statistical analysis: Analysis of variance (GLM procedure, Statistica, verion 6.1; StatSoft, Inc.) was used to determine significance of treatment effects and
interactions. Fisher's post-hoc least significant difference test was used to determine
significance of differences among means. Statistically significant differences are
defined at the 95% confidence interval. Data shown are means ± SEM.
RESULTS
Ajoene reduces cell viability: Adipocytes were treated with different
concentrations (0, 50, 100, 200, 400 μM) of ajoene for 6, 12, and 24 h. After
treatment, the number of live cells was determined by MTS assay. As shown in FIG. 1, ajoene time and dose-dependently reduced viability in adipocytes. Ajoene at 200
μM decreased cell viability about 50% after 24 h of treatment. These concentrations
of ajoene were selected for subsequent analyses.
Ajoene induces apoptosis: To investigate whether the reduction in cell number by ajoene was due to apoptosis, a ssDNA ELISA assay was used as a determinant of
cellular apoptosis. As shown in FIG. 2A, exposure of adipocytes to ajoene resulted in
a time- and dose-dependent induction of cell death that was detected after 6 h with
200 μM ajoene and higher. Longer (e.g., 24 h) exposures to ajoene significantly
increased apoptosis at 200 and 400 μM ajoene (p<0.05). DNA extracts from
adipocytes treated with ajoene displayed a characteristic apoptotic ladder pattern of
discontinuous DNA fragments on agarose gel electrophoresis (FIG. 2B).
Ajoene triggers apoptosis by oxidative stress: To determine the involvement of
ROS in ajoene-induced apoptosis, ROS levels were determined in ajoene-treated
adipocytes. As shown in FIG. 3A, ajoene increased ROS production by 2.1 fold after
5 min, reaching a plateau after 20 min. There was a decrease in ROS production between 20 and 40 minutes after treatment. Furthermore, when adipocytes were
pretreated with 10 mM NAC (a thiol-containing antioxidant) for 1 hr prior to
incubation with ajoene, ajoene-induced apoptosis was effectively reduced (FIG. 3B) to
nearly the control level. However, pretreatment of adipocytes with 400 units of
catalase (a scavenger for hydrogen peroxide) did not prevent the ajoene-induced
apoptosis.
Ajoene induces activation of MAPKs: Since the apoptotic effects of ajoene
may involve the generation of ROS (8) and some alterations in oxidative state are
intimately connected to perturbations in MAP kinase pathways (9), studies were
undertaken to determine the relationship between ajoene-induced oxidative stress and changes in JlSfK and ERKl/2 activation. Incubation of adipocytes with ajoene (200
μM) led to phosphorylation of JNK (FIG. 4A, upper panel). Quantitative analysis
(FIG. 4A) shows that activation of JNK occurred as late as 60 min with maximum
activity at 180 min, after which JNK decreased gradually. Interestingly, ERKl/2, a
kinase suggested to play a role in survival pathways, was also phosphorylated upon
exposure to ajoene. FIG. 4B (upper panel) shows the time-dependent phosphorylation
of ERKl/2 in adipocytes treated with ajoene (200 μM). Maximum activation of
ERKl/2 was observed 30 min after incubation with ajoene and quickly diminished
after 60 min. Pretreatment with the antioxidant NAC (10 niM) completely blocked
both ajoene-mediated ERKl/2 activation and JNK activation (FIGS. 4C and 4D).
Ajoene induces PARP-J cleavage and AIF-mediated cell death in a caspase-
independent manner: Since it has been shown that ROS-mediated DNA damage
triggers activation of PARP and subsequent cell death (18, 25), the role of PARP in
ajoene-induced apoptotic cell death was also investigated. Treatment of adipocytes
with 200 μM ajoene induced the proteolytic cleavage of PARP-I (116 kDa), resulting
in the accumulation of the 85 kDa cleavage product (FIG. 5 A, upper panel). Western
blotting and quantitative data revealed that PARP-I cleavage was apparent 4 hr after ajoene treatment and gradually increased. Moreover, pretreatment with the
antioxidant NAC blocked ajoene-mediated degradation of PARP-I (FIG. 5B). Surprisingly, ajoene did not activate caspase-3; either through proteolytic cleavage of
the proenzyme (FIG. 5A) or through protease activity (FIG. 5C). Taken together,
these data indicate that ROS-mediated cell death involved PARP degradation in a
caspase-independent manner.
Whether ajoene-induced cell death involved apoptosis-inducing factor (AIF)
was examined, because AJP-induced cell death is also caspase independent (17). The adipocytes exposed to ajoene (200 μM) showed a translocation of AIF from the
mitochondria to the nuclei in a time-dependent manner (FIG. 6A). Quantitative
analysis also showed that the amount of nuclear AIF gradually increased from 4 h to
12 h. Moreover, the pretreatment with the antioxidant NAC prevented translocation
of AIF to nuclei (FIG. 6B).
DISCUSSION
The present example elucidates the biological effect of ajoene, a component of
garlic, on 3T3-L1 adipocytes. These results represent the first insight into the mechanism of ajoene-induced apoptosis by ROS in 3T3-L1 adipocytes. ROS generation is first shown here to activate the MAPK cascade and PARP-I . These
results show that increased ROS production is involved in AIF release in a caspase-
independent manner.
To elucidate the mechanism of apoptosis by ajoene, MAPK expression and
ROS production were investigated. Results show that treatment with ajoene led to
phosphorylation of both ERK1/2 and JNK in 3T3-L1 adipocytes. In general, JNK
activation is associated with apoptosis induction, whereas ERK activation is
cytoprotective (14). Although it is generally accepted that activation of ERKl/2 leads
to cell proliferation (26), there are conditions in which ERKl/2 activation results in cell death (27, 28). Furthermore, previous studies have shown that H2O2 (ROS
stimulator) rapidly activated ERK in PC 12 cells (29), and ROS are involved in cell
death via the ERKl/2 signaling pathway (30). The present results also showed that
ROS levels peaked 20 min after ajoene treatment, whereas ERKl/2 was activated at 30 min and JNK was activated 180 min, which suggests that within this context, ROS
is likely to be upstream of MAPK activation. These data indicate that ajoene
differentially influences the phosphorylation status of members of the MAP kinase superfamily; the phosphorylation of ERK1/2 rapidly decreased, whereas the
phosphorylation of INK slowly and modestly decreased. In addition, the observation
that pre-treatment of cells with NAC inhibited both ROS generation and ERK1/2 and
JNK protein expression suggests that ROS directly influences MAPK signaling.
Catalase is a scavenger for hydrogen peroxide and did not block ajoene-induced ROS generation. NAC, however, is a thiol-reducing agent in addition to its action as a free
radical scavenger (31). Ajoene, which contains thiol groups, may be reduced by NAC and thereby lose its ability to induce apoptosis through generation of ROS and subsequent MAPK activation.
Some studies have implicated the members of the MAPK family as regulators
of mitochondrial-dependent apoptosis (32). A recent report showed that JNK up-
regulation is followed by the activation of Bax and down-stream mitochondrial
depolarization, with the release of AIF and cytochrome c, in a caspase-independent
manner (33). Therefore, these signaling pathways may be involved in the ROS-
induced mitochondrial changes by ajoene. The present results demonstrate that ajoene
did not affect the activity of caspase-3 either by proteolytic cleavage of the proenzyme
or by protease activity. This suggests that the apoptotic cell death induced by ajoene
treatment is caspase independent.
It has been reported that AIF mediates cell death through a caspase-
independent pathway. Death stimuli causes translocation of AJJF from the
mitochondria to the nucleus, where it initiates nuclear condensation (17, 34). Once
the nucleus condenses, this leads to large-scale chromatin fragmentation followed by
cell death. Consistent with these findings, translocation of AlF from mitochondria to
the nucleus and DNA fragmentation occurred. In addition, the treatment of cells with
NAC inhibited AIF translocation to the nucleus, suggesting that the increased intracellular ROS level is critical for the AIF relocalization after ajoene treatment.
These observations indicate that ajoene induces ROS generation followed by AIF
translocation and cell death, which can be reversed by preventing generation of ROS.
PARP is a nuclear enzyme that facilitates DNA repair in response to DNA
damage (35, 36). Enhanced activation of poly(ADP-ribose) polymerase (PARP)
enzyme is a major contributor to oxidative stress-induced cell dysfunction and tissue
injury (37, 38). Reactive oxygen species (ROS) cause single-strand DNA breaks (39).
Single-strand DNA breaks can activate nuclear PARP. IfPARP activation exceeds a certain limit, it can lead to cellular NAD+ and ATP depletion, ultimately resulting in
cell death (37-40). The present data also showed that an increased level of ROS by ajoene induced cleavage of 116 kDa PARP-I resulted in the accumulation of an 85
kDa product. Further treatment with NAC attenuated cleavage of PARP-I.
hi summary, these findings demonstrate that ajoene treatment induced
apoptosis through the caspase-independent cell death pathway by enhancement of
intracellular ROS level. The enhancement of intracellular ROS level promotes
MAPK and PARP-I activation and subsequent AIF release, ultimately resulting in
apoptosis. Thus, ajoene may be a new therapeutic tool for the treatment of obesity by regulating fat cell number through the induction of adipocyte apoptosis.
It should be emphasized that the above-described embodiments of the present
disclosure are merely possible examples of implementations, and are set forth only for
a clear understanding of the principles of the disclosure. Many variations and
modifications may be made to the above-described embodiments of the disclosure
without departing substantially from the spirit and principles of the disclosure. All
such modifications and variations are intended to be included herein within the scope
of the present disclosure and protected by the following claims. References:
Various publications are referenced parenthetically throughout this application,
the full citations for which are provided below. These publications are incorporated
by reference herein.
1. Moller DE and Flier JS. Insulin resistance—mechanisms, syndromes, and
implications. NEnglJMed. 1991;325:938-948.
2. Spiegelman BM, Choy L, Hotamisligil GS, Graves RA, and Tontonoz P. Regulation of adipocyte gene expression in differentiation and syndromes of
obesity/diabetes. J Biol Chem. 1993;268:6823-6826.
3. Hirsch J and Han PW. Cellularity of rat adipose tissue: effects of growth,
starvation, and obesity. J Lipid Res. 1969;10:77-82.
4. Prins JB and O'Rahilly S. Regulation of adipose cell number in man. Clin Sd
(Lond). 1997;92:3-11.
5. Siegers CP, Steffen B, Robke A, and Pentz R. The effects of garlic
preparations against human tumor cell proliferation. Phytomedicine. 1999;6:7-11.
6. Sigounas G, Hooker J, Anagnostou A, and Steiner M. S-allylmercaptocysteine
inhibits cell proliferation and reduces the viability of erythroleukemia, breast, and
prostate cancer cell lines. Nutr Cancer. 1997;27: 186-191.
7. Nishikawa T, Yamada N, Hattori A, Fukuda H, and Fujino T. Inhibition by
ajoene of skin-tumor promotion in mice. Biosci Biotechnol Biochem. 2002;66:2221-
2223.
8. Dirsch VM, Gerbes AL, and Vollmar AM. Ajoene, a compound of garlic,
induces apoptosis in human promyeloleukemic cells, accompanied by generation of reactive oxygen species and activation of nuclear factor kappaB. MoI Pharmacol.
1998;53:402-407.
9. Martindale JL and Holbrook NJ. Cellular response to oxidative stress:
signaling for suicide and survival. J Cell Physiol. 2002;192:l-15.
10. Chen YR, Wang W, Kong AN, and Tan TH. Molecular mechanisms of c-Jun
N-terminal kinase-mediated apoptosis induced by anticarcinogenic isothiocyanates. J
Biol Chem. 1998;273:1769-1775.
11. Shiah SG, Chuang SE, Chau YP, Shen SC, and Kuo ML. Activation of c-Jun
NH2-terminal kinase and subsequent CPP32/Yama during topoisomerase inhibitor beta-lapachone-induced apoptosis through an oxidation-dependent pathway. Cancer
Res. 1999;59:391-398.
12. Watabe M, Kakeya H, and Osada H. Requirement of protein kinase
(Krs/MST) activation for MT-21 -induced apoptosis. Oncogene. 1999;18:5211-5220.
13. Zhuang S, Demirs JT, and Kochevar IE. p38 mitogen-activated protein kinase
mediates bid cleavage, mitochondrial dysfunction, and caspase-3 activation during
apoptosis induced by singlet oxygen but not by hydrogen peroxide. J Biol Chem. 2000;275:25939-25948.
14. Ichijo H, Nishida E, Me K, et al. Induction of apoptosis by ASKl , a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways.
Science. 1997;275:90-94.
15. Szabados E, Fischer GM, Toth K, et al. Role of reactive oxygen species and
poly-ADP-ribose polymerase in the development of AZT-induced cardiomyopathy in
rat. Free Radic Biol Med. 1999;26:309-317. 16. Pacher P, Liaudet L, Mabley J, Komjati K, and Szabo C. Pharmacologic
inhibition of poly(adenosine diphosphate-ribose) polymerase may represent a novel
therapeutic approach in chronic heart failure. J Am Coll Cardiol. 2002;40: 1006-1016.
17. Susin SA, Lorenzo HK, Zamzami N, et al. Molecular characterization of
mitochondrial apoptosis-inducing factor. Nature. 1999;397:441-446.
18. Yu SW, Wang H, Poitras MF, et al. Mediation of poly(ADP-ribose)
polymerase-1 -dependent cell death by apoptosis-inducing factor. Science.
2002;297:259-263.
19. Park SY, Cho SJ, Kwon HC, Lee KR, Rhee DK, and Pyo S. Caspase- independent cell death by allicin in human epithelial carcinoma cells: involvement of
PKA. Cancer Lett. 2005 ;224: 123-132.
20. Hemati N, Ross SE, Erickson RL, Groblewski GE, and MacDougald OA.
Signaling pathways through which insulin regulates CCAAT/enhancer binding protein
alpha (C/EBPalpha) phosphorylation and gene expression in 3T3-L1 adipocytes.
Correlation with GLUT4 gene expression. J Biol Chem. 1997;272:25913-25919.
21. Frankfurt OS. Immunoassay for single-stranded DNA in apoptotic cells.
Methods MoI Biol. 2004;282:85-102.
22. Frankfurt OS and Krishan A. Identification of apoptotic cells by formamide-
induced dna denaturation in condensed chromatin. JHistochem Cytochem.
2001;49:369-378.
23. Bradford MM. A rapid and sensitive method for the quantitation of microgram
quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248-254.
24. Jacobson MD and Raff MC. Programmed cell death and Bcl-2 protection in
very low oxygen. Nature. 1995;374:814-816. 25. Althaus FR, Kleczkowska HE, Malanga M, et al. Poly ADP-ribosylation: a
DNA break signal mechanism. MoI Cell Biochem. 1999;193:5-11.
26. Grewal SS, York RD, and Stork PJ. Extracellular-signal-regulated kinase
signalling in neurons. Curr Opin Neurobiol. 1999;9:544-553.
27. Mohr S, McCormick TS, and Lapetina EG. Macrophages resistant to
endogenously generated nitric oxide-mediated apoptosis are hypersensitive to exogenously added nitric oxide donors: dichotomous apoptotic response independent
of caspase 3 and reversal by the mitogen-activated protein kinase kinase (MEK) inhibitor PD 098059. Proc Natl Acad Sd USA. 1998;95:5045-5050.
28. Murray B5 Alessandrini A, Cole AJ, Yee AG3 and Furshpan EJ. Inhibition of
the p44/42 MAP kinase pathway protects hippocampal neurons in a cell-culture model
of seizure activity. Proc Natl AcadSci USA. 1998;95:11975-11980.
29. Zhang L and Jope RS. Oxidative stress differentially modulates
phosphorylation of ERK, p38 and CREB induced by NGF or EGF in PC12 cells.
Neurobiol Aging. 1999;20:271-278.
30. Rothstein EC, Byron KL, Reed RE, Fliegel L, and Lucchesi PA. H(2)O(2)-
induced Ca(2+) overload in NRVM involves ERKl/2 MAP kinases: role for an NHE-
1-dependent pathway. Am J Physiol Heart Circ Physiol. 2002;283:H598-605.
31. Arakaki N, Kajihara T, Arakaki R, et al. Involvement of oxidative stress in
tumor cytotoxic activity of hepatocyte growth factor/scatter factor. J Biol Chem.
1999;274:13541-13546.
32. Ghatan S, Lamer S, Kinoshita Y, et al. p38 MAP kinase mediates bax
translocation in nitric oxide-induced apoptosis in neurons. J Cell Biol. 2000,150:335-
347. 33. Boisvieux-Ulrich E, Sourdeval M, and Marano F. CD437, a synthetic retinoid,
induces apoptosis in human respiratory epithelial cells via caspase-independent
mitochondrial and caspase-8-dependent pathways both up-regulated by INK signaling
pathway. Exp Cell Res. 2005;307:76-90.
34. Ferri KF, Jacotot E, Blanco J, et al. Apoptosis control in syncytia induced by
the HIV type 1-envelope glycoprotein complex: role of mitochondria and caspases. J
Exp Med. 2000;192:1081-1092.
35. Lautier D, Lagueux J, Thibodeau J, Menard L, and Poirier GG. Molecular and biochemical features of poly (ADP-ribose) metabolism. MoI Cell Biochem.
1993;122:171-193.
36. de Murcia G, Schreiber V, Molinete M, et al. Structure and function of
poly(ADP-ribose) polymerase. MoI Cell Biochem. 1994;138:15-24.
37. Virag L and Szabo C. The therapeutic potential of poly( ADP-ribose)
polymerase inhibitors. Pharmacol Rev. 2002;54:375-429.
38. Szabo G, Liaudet L, Hagl S, and Szabo C. Poly(ADP-ribose) polymerase
activation in the reperfused myocardium. Cardiovasc Res. 2004;61 :471-480.
39. Halmosi R, Berente Z, Osz E, Toth K, Literati-Nagy P, and Sumegi B. Effect
of poly( ADP-ribose) polymerase inhibitors on the ischemia-reperfusion-induced
oxidative cell damage and mitochondrial metabolism in Langendorff heart perfusion
system. MoI Pharmacol. 2001;59:1497-1505.
40. Habon T, Szabados E, Kesmarky G, et al. The effect of carvedilol on enhanced
ADP-ribosylation and red blood cell membrane damage caused by free radicals.
Cardiovasc Res. 2001;52:153-160.

Claims

CLAIMS The following is claimed:
1. A method of treating obesity comprising administering to a host in need of
treatment an effective amount of a compound selected from: ajoene,
precursors thereof, and derivatives thereof, wherein the compound is present in
a dosage level effective to initiate the release or activation of an apoptosis
inducing factor, wherein the apoptosis inducing factor leads to the apoptotic cell death of adipose tissue cells in the host.
2. A method for inducing apoptosis in adipose tissue cells in a host, comprising
initiating the release or activation of an apoptosis inducing factor by
administering an effective amount of at least one garlic extract compound,
wherein the apoptosis inducing factor leads to apoptotic cell death.
3. The method of claim 2, wherein the garlic extract compound is selected from
garlic thiosulfmates and transformation products thereof, ajoene, precursors thereof, and derivatives thereof; allicin, precursors thereof, and derivatives
thereof; allyl methanethiosulfinate, precursors thereof, and derivatives thereof;
disulfides, precursors thereof, and derivatives thereof; allylsulfides, precursors
thereof, and derivatives thereof; vinyldithiins, precursors thereof, and
derivatives thereof; diallyl trisulfides, precursors thereof, and derivatives
thereof; and mercaptocysteines, precursors thereof, and derivatives thereof;
and combinations thereof.
4. The method of claim 2, wherein the garlic extract compound is selected from
ajoene, precursors thereof, and derivatives thereof.
5. The method of claim 4, wherein administering an effective amount of at least
one garlic extract compound does not activate caspases.
6. The method of claim 4, wherein the apoptotic cell death occurs independently from caspases.
7. The method of claim 2, wherein the apoptosis inducing factor is Apoptosis
Inducing Factor (AIF).
8. The method of claim 2, wherein the apoptosis inducing factor is a mitogen-
activated protein kinase (MAPK).
9. The method of claim 8, wherein the MAPK is extracellular signal-regulating kinase (ERK1/2).
10. The method of claim 8, wherein the MAPK is Jun-N-terrninal kinase (INK).
11. The method of claim 2, wherein the apoptosis inducing factor is PoIy(ADP
ribose) polymerase (PAPvP).
12. A pharmaceutical composition comprising at least one garlic extract
compound in combination with a pharmaceutically acceptable carrier, wherein the at least one garlic extract compound is present in a dosage level effective
to initiate the release of an apoptosis inducing factor, wherein the apoptosis
inducing factor leads to the apoptotic cell death of adipose tissue cells in a
host.
13. The pharmaceutical composition of claim 12, wherein the apoptotic cell death of adipose tissue cells treats obesity.
14. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from garlic thiosulfinates and transformation products
thereof, ajoene, precursors thereof, and derivatives thereof; allicin, precursors
thereof, and derivatives thereof; allyl methanethiosulfmate, precursors thereof,
and derivatives thereof; disulfides, precursors thereof, and derivatives thereof;
allylsulfides, precursors thereof, and derivatives thereof; vinyldithiins,
precursors thereof, and derivatives thereof; diallyl trisulfides, precursors
thereof, and derivatives thereof; and mercaptocysteines, precursors thereof, and derivatives thereof; and combinations thereof.
15. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from ajoene, precursors thereof, and derivatives thereof.
16. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from garlic thiosulfinates and transformation products thereof.
17. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from allicin, precursors thereof, and derivatives thereof.
18. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from allyl methanethiosulfinate, precursors thereof, and
derivatives thereof.
19. The pharmaceutical composition of claim 12, wherein the garlic extract compound is selected from disulfides, precursors thereof, and derivatives
thereof.
20. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from allylsulfides, precursors thereof, and derivatives
thereof.
21. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from vinyldithiins, precursors thereof, and derivatives
thereof.
22. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from diallyl trisulfides, precursors thereof, and
derivatives thereof.
23. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from mercaptocysteines, precursors thereof, and
derivatives thereof.
24. The pharmaceutical composition of claim 12, wherein the garlic extract
compound is selected from S-allylmercaptocysteine (SAMC), S-
methylcysteine (SMC), S-allyl cysteine sulfoxide (SACS), diallyl disulfide (DADS), S-allylcysteine (SAC), S-ethylcysteine (SEC), S-ρropylcysteine
(SPC), and combinations thereof.
25. The pharmaceutical composition of claim 12, wherein the garlic extract is
selected from ajoene, precursors of thereof, and derivatives thereof, wherein
the dosage level of the ajoene, precursors of thereof, and derivatives thereof,
does not activate caspases.
26. The pharmaceutical composition of claim 12, wherein the apoptotic cell death of adipose tissue cells in the host occurs independently from caspases.
27. A method for treating osteoporosis in a host, comprising administering an
effective amount of at least one garlic extract compound.
28. The method of claim 27, wherein the garlic extract compound is selected from
ajoene, precursors thereof, and derivatives thereof.
29. A pharmaceutical composition comprising at least one garlic extract
compound in combination with a pharmaceutically acceptable carrier, wherein
the at least one garlic extract compound is present in a dosage level effective
to treat osteoporosis.
30. The pharmaceutical composition of claim 29, wherein the garlic extract
compound is selected from ajoene, precursors thereof, and derivatives thereof.
PCT/US2005/034632 2004-09-30 2005-09-29 Compositions and methods for inducing adipose tissue cell death WO2006039285A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61473804P 2004-09-30 2004-09-30
US60/614,738 2004-09-30

Publications (2)

Publication Number Publication Date
WO2006039285A2 true WO2006039285A2 (en) 2006-04-13
WO2006039285A3 WO2006039285A3 (en) 2006-06-29

Family

ID=36143004

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/034632 WO2006039285A2 (en) 2004-09-30 2005-09-29 Compositions and methods for inducing adipose tissue cell death

Country Status (1)

Country Link
WO (1) WO2006039285A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008053487A2 (en) 2006-11-01 2008-05-08 The Medical Research Fund At The Tel-Aviv Sourasky Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050003026A1 (en) * 2002-08-14 2005-01-06 Song-Hae Bok Powder or extracts of plant leaves with anti-obesity effects and anti-obesity food comprising them
US20050013921A1 (en) * 2003-07-17 2005-01-20 Peticca Andrea Lynn Novel multi-component garlic food supplement
US6852343B2 (en) * 2000-10-31 2005-02-08 Pierre Fabre Dermo-Cosmetique Antiadipose topical treatment composition based on garlic bulbs extracts, and cosmetic and therapeutic uses

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6852343B2 (en) * 2000-10-31 2005-02-08 Pierre Fabre Dermo-Cosmetique Antiadipose topical treatment composition based on garlic bulbs extracts, and cosmetic and therapeutic uses
US20050003026A1 (en) * 2002-08-14 2005-01-06 Song-Hae Bok Powder or extracts of plant leaves with anti-obesity effects and anti-obesity food comprising them
US20050013921A1 (en) * 2003-07-17 2005-01-20 Peticca Andrea Lynn Novel multi-component garlic food supplement

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NELSON-DOOLEY C.: 'Novel Treatments for Obesity and Osteoporosis: Targeting Apoptotic Pathways in Adipocytes' CURRENT MEDICINAL CHEMISTRY vol. 12, no. 19, September 2005, pages 2215 - 2225, XP008063180 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008053487A2 (en) 2006-11-01 2008-05-08 The Medical Research Fund At The Tel-Aviv Sourasky Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
EP2078079B1 (en) * 2006-11-01 2011-05-04 The Medical Research and Infrastructure Fund of the Tel-Aviv Sourasky Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US9279127B2 (en) 2006-11-01 2016-03-08 The Medical Research Fund At The Tel-Aviv Sourasky Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression
US9663790B2 (en) 2006-11-01 2017-05-30 The Medical Research, Infrastructure, And Health Services Fund Of The Tel Aviv Medical Center Adipocyte-specific constructs and methods for inhibiting platelet-type 12 lipoxygenase expression

Also Published As

Publication number Publication date
WO2006039285A3 (en) 2006-06-29

Similar Documents

Publication Publication Date Title
US8071795B2 (en) HIF inhibitors
KR101221505B1 (en) New use for cannabinoid
WO2018140599A1 (en) Benzothiophene and benzothiazole compounds and methods of use thereof
EP3573970A1 (en) 1-(6-(3-hydroxynaphthalen-1-yl)quinazolin-2-yl)azetidin-1-yl)prop-2-en-1-one derivatives and similar compounds as kras g12c inhibitors for the treatment of cancer
EP3573954A1 (en) Fused bicyclic benzoheteroaromatic compounds and methods of use thereof
US9504674B2 (en) TrkB agonists and methods of use
KR20070089151A (en) New use for cannabinoid
RU2715419C1 (en) Pharmaceutical compositions and methods of treating cardiovascular diseases
US11413278B2 (en) Compounds and methods for promoting stress resistance
TW201722897A (en) Compounds effective in treating hepatotoxicity and fatty liver diseases and uses thereof
US11524055B2 (en) Methods for treating diseases mediated by ERBB4-positive pro-inflammatory macrophages
JP7395480B2 (en) Pharmaceutical compositions containing phenylsulfonamides and their therapeutic applications
WO2004103057A2 (en) Compositions and methods for inducing adipose tissue cell death
WO2006039285A2 (en) Compositions and methods for inducing adipose tissue cell death
JP2019514843A (en) Uridines phosphoramide prodrugs, process for their preparation and their use in medicine
JP2011522039A (en) Diarylhepatonoid compounds useful as virus inhibitors
US20030171379A1 (en) Methods of treating, preventing, or inhibiting inflammation with Mactanamide compounds
WO2007089685A2 (en) Compositions and methods for inducing adipose tissue cell death
US20150174112A1 (en) Compositions for treating fragile x and other disorders methods of use thereof, and screening for compounds for fragile x and other disorders
WO2005091804A2 (en) Paramyxovirus family inhibitors and methods of use thereof
US8618077B2 (en) Compositions and methods for treating fragile X premutation RVGG repeats-mediated toxicity
US11622960B2 (en) Microtubule polymerization inhibitor prodrugs and methods of using the same
WO2018170509A1 (en) Fatty acid synthase inhibitors and methods of use
Lee et al. Suppressive activities of KC1–3 on HMGB1-mediated septic responses
US11370743B2 (en) Prodrug derivatives of protein kinase C modulators

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05801030

Country of ref document: EP

Kind code of ref document: A2