US20090082433A1 - Preventative treatment and remission of allergic diseases - Google Patents

Preventative treatment and remission of allergic diseases Download PDF

Info

Publication number
US20090082433A1
US20090082433A1 US12/281,517 US28151707A US2009082433A1 US 20090082433 A1 US20090082433 A1 US 20090082433A1 US 28151707 A US28151707 A US 28151707A US 2009082433 A1 US2009082433 A1 US 2009082433A1
Authority
US
United States
Prior art keywords
plant
extract
kaempferia
ether
organic solvent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/281,517
Other languages
English (en)
Inventor
Roelof Marthinus Horak
Ebrahim Wadiwala
Gerda Fouche
Marina Mikhailovna Van Der Merwe
Vinesh Jaichand Maharaj
Louis Gabriel Jozua Ackerman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Council for Scientific and Industrial Research CSIR
Original Assignee
Council for Scientific and Industrial Research CSIR
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 Council for Scientific and Industrial Research CSIR filed Critical Council for Scientific and Industrial Research CSIR
Assigned to CSIR reassignment CSIR ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ACKERMAN, LOUIS GABRIEL JOZUA, FOUCHE, GERDA, HORAK, ROELOF MARTHINUS, MAHARAJ, VINESH JAICHAND, VAN DER MERWE, MARINA MIKHAILOVNA, WADIWALA, EBRAHIM
Publication of US20090082433A1 publication Critical patent/US20090082433A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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)
    • A61K36/906Zingiberaceae (Ginger family)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents

Definitions

  • THIS INVENTION relates to compounds useful in the preventative treatment and remission of allergic diseases. More specifically, this invention relates to compounds useful in the preventative treatment and remission of asthma and/or atopy.
  • Glucocorticoids are widely used in the preventative treatment of asthma and other allergic diseases. Glucocorticoids bind to the glucocorticoid receptor (GR), thereby activating the GR. The activated GR then binds to a glucocorticoid responsive element and upregulates the genes for anti-inflammatory agents such as lipocortin. The induction of the anti-inflammatory protein lipocortin in turn inhibits the enzyme phospholipase A2, thereby decreasing the production of allergy mediators, such as prostaglandins and leukotrienes. Allergy and Allergic Diseases: The new Mechanism and Therapeutics , edited by J. A.
  • cytokines and chemokine agents such as Interleukin-4 (IL-4) and Interleukin-5 (IL-5).
  • IL-4 Interleukin-4
  • IL-5 Interleukin-5
  • PDE phosphodiesterase isoenzymes
  • PDE's phosphodiesterase isoenzymes
  • PDE 4 phosphodiesterase isoenzymes
  • Leukotrienes are also of particular interest in the study of allergic disease, because of their marked bronchoconstrictory action. Leukotrienes have an approximate 1000-fold greater bronchoconstrictory action than histamines and prostaglandins. Leukotrienes are by-products of arachidonic acid which is located in phospholipid bi-layers in the cell membranes of mast cells. During an asthma attack, arachidonic acid is converted into five leukotrienes.
  • leukotriene biosynthesis inhibitors have an important role to play, since they inhibit the action of 5-LO, thereby inhibiting the eventual synthesis of the bronchoconstrictory leukotrienes.
  • the above pathway may thus be manipulated by administering a glucocorticoid receptor (GR) binding compound, a PDE inhibitor and/or a 5-LO inhibitor to a patient in need thereof.
  • GR glucocorticoid receptor
  • PDE glucocorticoid receptor
  • 5-LO 5-LO inhibitor
  • NF- ⁇ B nuclear factor-KB
  • NF- ⁇ B is a central mediator of the human immune response, regulating the transcription of various pro-inflammatory and inflammatory mediators such as the cytokines Interleukin-1 (IL-1), Interleukin-2 (IL-2), Interleukin-8 (IL-8) and TNF- ⁇ , as well as genes encoding cyclo-oxygenase II, nitric oxide synthase, immunoreceptors, cell adhesion molecules, or acute phase proteins.
  • IL-1 Interleukin-1
  • IL-2 Interleukin-2
  • IL-8 Interleukin-8
  • TNF- ⁇ TNF- ⁇
  • ICAM -1 expression is highly NF - kappaB - dependent in A 549 cells. European Journal of Biochemistry. 271: 785-791. Therefore, inhibition of NF- ⁇ B resulting in the down regulation of chemokines such as IL-8 could be beneficial in the treatment of asthma and inflammatory diseases.
  • an organic solvent extract or an essential oil of a plant of the family Zingiberaceae for use in the preventative treatment and remission of allergic diseases.
  • the plants of the family Zingiberaceae include the genera Siphonochilus, Kaempferia, Cienkowskia and/or Cienkowskiella.
  • the organic solvent extract or essential oil may be obtained from plant material of plants selected from the species Siphonochilus aethiopicus, Siphonochilus natalensis, Kaempferia aethiopica, Kaempferia natalensis, Kaempferia ethelae, Cienkowskia aethiopica and Cienkowskiella aethiopica.
  • the said organic solvent extract or essential oil comprises as an active ingredient a compound having the structural formula 1:
  • the chemical name (IUPAC) of the compound of formula 1 is 4,4a,5,9-tetrahydro-3,5,8a-trimethylnaptho[2,3-b]furan-8-one.
  • the compound of formula 1 may be used in the form of a racemic mixture or in the form of one of its steroisomers.
  • allergic diseases examples include asthma and atopy.
  • the organic solvent extract or essential oil is capable of binding to a glucocorticoid receptor (GR), inducing the production of lipocortin, and/or the inhibition of the enzyme phospholipase A2, and/or the decreasing of production of prostaglandins and/or leukotrienes.
  • GR glucocorticoid receptor
  • the organic solvent extract or essential oil may have anti-bronchoconstrictory activity and further may downregulate cytokines and chemokine agents such as Interleukin-4 (IL-4) and Interleukin-5 (IL-5).
  • IL-4 Interleukin-4
  • IL-5 Interleukin-5
  • the organic solvent extract or essential oil may inhibit NF- ⁇ B a central mediator of the human immune response, down regulating the transcription of various pro-inflammatory and inflammatory mediators such as the cytokines Interleukin-8 (IL-8)
  • IL-8 Interleukin-8
  • the organic solvent extract or essential oil may be obtainable by a method which includes the steps of preparing an extract from plant material of a plant of the family Zingiberaceae, and separating a fraction having activity against allergic diseases, the extracts containing the active ingredient having activity against allergic diseases.
  • the method may include the steps of extracting wet rhizomes or roots or ground plant samples of said plant species such as Siphonochilus aethiopicus , or drying the rhizomes and/or the roots of the plant by air drying or oven drying, followed by grinding of the rhizomes and/or roots to a powder.
  • Extracts are prepared by extraction using organic solvents such as diethyl ether, di-isopropyl ether, t-butyl methyl ether, t-butyl ethyl ether, ethyl acetate or benzyl acetate.
  • the active ingredient may be extracted by extraction techniques which include steam distillation, and or purification of the extracts using solvent/solvent partitioning and/or chromatographic separating techniques.
  • a substance or composition for use in a method for the preventative treatment and remission of allergic diseases which substance or composition includes as an active ingredient an extract as described above, and said method comprising administering to a subject an effective dosage of said substance or composition.
  • a substance or composition for use in a method for the preventative treatment and remission of allergic diseases which substance or composition includes as an active ingredient a compound of formula 1, and said method comprising administering to a subject an effective dosage of said substance or composition.
  • a compound of formula 1 for use in a method for the preventative treatment and remission of allergic diseases.
  • allergic diseases examples include asthma and atopy.
  • composition for use in the preventative treatment and remission of allergic diseases which includes an effective quantity of one of said extract, said compound of formula 1 or said essential oil.
  • the extract, and compositions of the extract may be in a form suitable for administering to mammalian subjects, particularly human subjects.
  • the extract may be in the form of an organic solvent extract and/or an essential oil or combinations thereof.
  • the invention extends to the use of the organic solvent extract as described above, the essential oil or the compound of formula 1 in the downregulation of glucocorticoid receptors, the inhibition of phospholipase A2, the downregulation of allergy mediators such as prostaglandins and leukotrienes, the downregulation of cytokines such as IL-4 and IL-5, the inhibition of phosphodiesterase 4, the inhibition of 5-lipoxygenase or leukotriene biosynthesis, and the inhibition of specific activity of the NF- ⁇ B Transcription Response resulting in down regulation of IL-8.
  • the invention provides the use of an extract of at least one plant selected from plants of the family Zingiberaceae in the preparation of a medicament for use in the treatment or prophylaxis of allergic diseases.
  • the at least one plant may be selected from the genera Siphonochilus, Kaempferia, Cienkowskia and Cienkowskiella and from the species Siphonochilus aethiopicus, Siphonochilus natalensis, Kaempferia aethiopica, Kaempferia natalensis, Kaempferia ethelae, Cienkowskia aethiopica and Cienkowskiella aethiopica.
  • the extract may include, as an active ingredient, a compound selected from compounds having the structural formula 1,
  • the allergic disease may be selected from asthma and atopy.
  • the extract may be an essential oil obtained by steam distillation of plant material from the at least one plant.
  • the extract may be an organic solvent extract obtained by extraction of plant material of the at least one plant with an organic solvent.
  • the organic solvent may be an ether selected from diethyl ether, diisopropyl ether, t-butyl methyl ether, t-amyl methyl ether and t-butyl ethyl ether.
  • the organic solvent may be an ester selected from methyl acetate, ethyl acetate and benzyl acetate.
  • the plant material may be obtained from roots or rhizomes of the plant.
  • the invention extends to a composition for use in a method of treatment or prophylaxis of allergic diseases, the composition including an extract of at least one plant selected from plants of the family Zingiberaceae.
  • the at least one plant may be as hereinbefore described.
  • the extract may be as hereinbefore described.
  • the allergic disease may be selected from asthma and atopy and the plant material may be obtained from roots or rhizomes of the plant.
  • the invention extends to the use of an extract of at least one plant selected from plants of the family Zingiberaceae in the preparation of a medicament for use in any one or more of the downregulation of glucocorticoid receptors, the inhibition of phospholipase A2, the downregulation of allergy mediators, the downregulation of cytokines, the inhibition of phosphodiesterase 4, the inhibition of 5-lipoxygenase or leukotriene biosynthesis, and the inhibition of specific activity of the NF- ⁇ B Transcription Response.
  • the allergy mediators may be selected from prostaglandins and leukotrienes.
  • the cytokines may be selected from IL-4, IL-5 and IL-8.
  • the at least one plant may be as hereinbefore described.
  • the extract may be as hereinbefore described.
  • the allergic disease may be selected from asthma and atopy.
  • the plant material may be obtained from roots or rhizomes of the plant.
  • the invention extends, further, to the use of a compound selected from compounds having the structural formula 1,
  • stereoisomers thereof and mixtures of stereoisomers thereof in the preparation of a medicament for use in the treatment or prophylaxis of allergic diseases are included in the preparation of a medicament for use in the treatment or prophylaxis of allergic diseases.
  • the allergic disease may be selected from asthma and atopy.
  • the invention extends further to a method of treatment of an allergic disease, the method including administering to a person or animal in need of treatment a therapeutically effective amount of an extract, a composition or a compound selected from compounds of structural formula 1 as hereinbefore described.
  • the allergic disease may be as hereinbefore described.
  • FIG. 1 shows a flow diagram illustrating a protocol for the extraction of the compound of formula 1
  • FIG. 2 shows an inhibition curve for Con-A induced cytotoxicity mediator release by the compound of formula 1;
  • FIG. 3 shows an inhibition curve for Con-A induced cytotoxicity mediator release by a DMSO standard control
  • FIG. 4 shows an inhibition or concentration response curve for NF- ⁇ B Transcription Response for the compound of formula 1, in which the curve has been normalised;
  • FIG. 5 shows an inhibition or concentration response curve for NF- ⁇ B Transcription Response for a Cyclosporin A positive control.
  • FIG. 6 shows a combined inhibition curve for IL-1 mediator release by both the compound of formula 1, as well as a Dexamethasone standard control
  • FIG. 7 shows a combined inhibition curve for IL-5 mediator release by both the compound of formula 1, as well as a Dexamethasone standard control, in which the inhibition curve of the compound of formula 1 has been normalised;
  • FIG. 8 shows an inhibition or concentration response curve for NF- ⁇ B Transcription Response for a diethyl ether extract containing the compound of formula 1, in which the curve has been normalised
  • FIG. 9 shows an inhibition or concentration response curve for NF- ⁇ B Transcription Response for a Cyclosporin A positive control.
  • the plant extract or distillate of the invention may be produced by extraction from wet rhizomes, roots or ground plant samples of Siphonochilus aethiopicus , or drying the rhizomes and/or the roots of the plant by air drying or oven drying, followed by grinding of the rhizomes and/or roots to a powder.
  • the active ingredient thereof may be extracted by extraction techniques which include steam distillation and solvent extraction.
  • the compound may be refined further by separating individual compounds from the extract or distillate using solvent/solvent partitioning and/or chromatographic separating techniques. These techniques are discussed in more detail in the illustrative Examples below.
  • an infusion 14 is prepared from plant material such as leaves, rhizomes or roots.
  • plant material such as leaves, rhizomes or roots.
  • One litre of deionised boiling water, shown at 12 is added to 22.08 g of oven dried (60° C.) ground rhizomes, shown at 10, and left to stand for 1 hour with occasional stirring.
  • the water is filtered and solids 11 are removed and the aqueous phase is extracted four times with 500 ml of diethyl ether, as shown at 16.
  • the extracts obtained from each extraction are combined, dried, filtered and the solvent removed by means of a rotary evaporator in a water bath at a temperature of 25° C., as shown at 18, to produce an ether portion 20 (WG13A) weighing 119 mg, which is designated the organic fraction.
  • An intermediate layer in the form of a solid 22 (WG13D) forms during the liquid-liquid partitioning. Approximately 107 mg of solid 22 is obtained, which is kept separate.
  • the aqueous layer is freeze-dried, as shown at 24, to yield 2.52 g of solid material 26 (WG13B) which is designated the aqueous fraction.
  • the compound of formula 1 is present in the organic fraction and is identified using thin layer chromatography. The compound may then be purified further using column chromatography.
  • one litre of diethyl ether is added to 22.08 g of oven dried (60° C.) ground rhizomes and left to stand for 1 hour with occasional stirring.
  • the ether is filtered and carefully evaporated under low vacuum to yield a dry extract 34 (WG-I-101 or WG-1-58A) of approximately 1.5 g.
  • the compound of formula 1 is identified in the organic extract by using thin layer chromatography.
  • the diethyl ether extracts produced by either the aqueous extraction or the organic extraction procedures described above was purified further by flash chromatography (silica gel), as shown at 30, using ethyl acetate-hexane (1:9, v/v) as eluent to yield the sesquiterpenoid of formula 1 WG13C or WG-I-94B, as shown at 32.
  • An essential oil is obtained by steam distillation of the rhizomes and/or roots of the plant species Siphonochilus aethiopicus .
  • the chemical compositions of the major components of the essential oil are shown in Table 1 below. Identification of the major components is accomplished by using combined gas chromatography-mass spectrometry and the Kovats Indices technique.
  • the sesquiterpenoid of formula 1 crystallizes at the interface of the essential oil and the condensate obtained during the steam distillation process. Upon subsequent cooling of the essential oil to below 4° C., the compound of formula 1 crystallizes from the oil.
  • the mixture of the essential oil and water in the separating funnel condensate is allowed to stand at room temperature for 16 h.
  • the resulting crystals present on the surface, comprising the compound of formula 1 are removed by draining of the condensate and filtering using a sintered glass funnel. A total of 480 mg of the compound of formula 1 is thus obtained.
  • the structural assignment of the compound of formula 1 is based on a detailed study of the high field 1 H and 13 C nuclear magnetic resonance (NMR) spectral data of the compound.
  • the first stage in the characterisation of the compound is the identification of the 1 H NMR signals belonging to isolated spin systems. This is achieved by means of two-dimensional (2D) ( 1 H, 1 H) correlation spectroscopy, using a COSY-45 sequence.
  • the multiplicities of the different resonances in the 13 C NMR spectra are deduced from the coupled 13 C NMR spectra as well from the proton decoupled CH, CH 2 , and CH 3 sub-spectra obtained using a DEPT pulse sequence.
  • the 13 C resonances are partly assigned by correlation of the proton-bearing carbon atoms, with specific resonances in 2D ( 13 C, 1 H) chemical-shift experiments.
  • Biochemical assay results as presented herein are reflected as the percent inhibition of specific binding or activity of a compound.
  • the assay is described in Cidlowski, J. A. and Cidlowski, N. B., Regulation of glucocorticoid receptors by glucocorticoids in cultured HeLa S 3 cells . Endocrinology 109: 1975-1982, 1981, the contents of which are incorporated herein by way of reference.
  • This assay specifically measures the ability of active glucocorticoids in glucocorticoid receptor downregulation. Responsive test compounds are therefore deemed to possess activities similar to those of active glucocorticoids.
  • Table 6 shows the results obtained from the screening of a combination of the organic ether fraction (designated WG-I-13A in FIG. 1 ), the aqueous fraction (designated WG-I-13D in FIG. 1 ) and the insoluble fraction (designated WG-I-13D in FIG. 1 ).
  • the combined fraction is designated WG-I-5AB+WG-I-5C, as shown in FIG. 1 .
  • glucocorticoid receptor (GR) binding assay (61% inhibition) in HeLa S 3 (human epitheloid cervic carcinoma) cells at a concentration of 100 ⁇ g/ml of the combined fraction (WG-I-5AB+WG-I-5C) concentration.
  • This assay measures binding of [ 3 H]Dexamethasone to human glucocorticoid receptors.
  • HeLa S3 cells are suspended in modified HEPES buffer, pH 7.2, using standard techniques.
  • the cells (1 ⁇ 10 6 ) are incubated with 6 nM [ 3 H]Dexamethasone for 120 minutes at 25° C. Non-specific binding is estimated in the presence of 20 ⁇ M Dexamethasone. Membranes are filtered and washed three times and the filters are counted to determine [ 3 H]Dexamethasone specifically bound. Compounds are screened at 10 ⁇ M for activity.
  • test compound and/or vehicle is incubated with 0.2 ⁇ g enzyme and 1 ⁇ M cAMP containing 0.01 ⁇ M [ 3 H]CAMP in Tris buffer pH 7.5 for 20 minutes at 30° C.
  • the reaction is terminated by boiling for 2 minutes and the resulting AMP is converted to adenosine by addition of 10 mg/ml snake venom necleotidase and further incubation at 30° C. for 10 minutes.
  • Unhydrolyzed cAMP is bound to AGI-X2 resin, and remaining [ 3 H]adenosine in the aqueous phase is quantified by scintillation counting.
  • Compounds are screened at 100 ⁇ M.
  • IBMX (3-isobutyl-1-methylxanthine) is used as standard reference agent.
  • the organic ether fraction (WG-I-13A), the aqueous fraction (WG-I-13B), as well as the insoluble fraction which forms at the interface of the organic and extract layer (designated WG-I-13D in FIG. 1 ), are separately bio-assayed using the glucocorticoid receptor (GR) binding assay and the phosphodiesterase PDE4 enzyme assay described above.
  • GR glucocorticoid receptor
  • the IC 50 value of the sesquiterpenoid compound in the glucocorticoid-binding assay was determined as being 50.3 ⁇ M, as shown in Table 11 below.
  • the test compound is incubated with Concanavalin A (ConA) (10 ⁇ g/ml)-stimulated human peripheral blood mononuclear leukocyte (PBMNL) cells in growth medium RPMI-1640 (pH 7.4) overnight at 37° C. in an incubator.
  • ConA Concanavalin A
  • PBMNL peripheral blood mononuclear leukocyte
  • RPMI-1640 pH 7.4
  • the IL-5 cytokine production levels in the conditioned medium are quantified using a sandwich ELISA kit.
  • Compounds are screened at 10, 1, 0.1, 0.01 and 0.001 ⁇ M. These same concentrations are concurrently applied to a separate group of treated cells and evaluated for possible compound-induced cytotoxicity only if significant inhibition of release is observed.
  • Dexamethasone is used as standard reference agent.
  • the test compound and/or vehicle is incubated with a suspension of human peripheral blood mononuclear leukocytes (PBMNL, 1 ⁇ 10 5 /well) in the presence of Concanavalin A (Con A, 20 ⁇ g/ml) in RPMI buffer (pH 7.4) at 37° C. overnight in 5% CO 2 .
  • AlamarBlue reagent is added and the cells are incubated at 37° C. for 16 hours. Living cells take up AlamarBlue and emit fluorescence. Fluorescence intensity is measured using a SpectraFluor Plus plate reader with excitation at 530 nm and emission at 590 nm. A decrease of 50% or more (50%) in fluorescence intensity relative to the vehicle treated controls indicates significant cytotoxicity.
  • Compounds are screened at 10, 1, 0.1, 0.01 and 0.001 ⁇ M. Dexamethasone is used as standard reference agent.
  • the IC 50 of the compound is 16.1 ⁇ M for the mediator release Interleukin-5 (IL-5) inhibition assay. Inhibition of 93% is observed for the mediator release Interleukin-5 (IL-5) inhibition assay, using 100 ⁇ M concentrations of the active compound in each experiment.
  • cytotoxicity of the active compound was observed when conducting the above bio-assays, as shown in Table 14, see IC 50 vs EC 50 values. Although a partial cytotoxic effect was observed during the mediator release assays, the efficacy of the purified compound of formula 1 is ascribed to the fact that effective binding to the glucocorticoid receptor (GR) site was established.
  • GR glucocorticoid receptor
  • An essential oil obtained by steam distillation of the dried rhizomes as described in Example 2, was also screened using the 5-lipoxygenase enzyme (5-LO) assay and the phosphodiesterase PDE4 assay to determine the inhibitory effect(s) thereof. Inhibition of 99% was observed using the 5-LO assay, while 73% inhibition was observed using the phosphodiesterase PDE4 assay, indicating that the essential oil has a bronchodilatory effect and may be used to inhibit the action of the 5-lipoxygenase enzyme. The results obtained are also shown in Table 12 above.
  • a diethyl ether extract was prepared in a manner similar to Example 1.
  • the rhizomes of the plant, Siphonochilus aethiopicus were washed, cut, oven dried at 40° C. and thereafter ground to a powder.
  • 1 of deionised boiling water was added to 25.0 g of oven dried ground rhizomes and left to stand for 1 hour with occasional stirring.
  • the water was filtered and extracted with diethyl ether (4 ⁇ 500 ml).
  • the ether layers were separated from the water layers, combined, dried (MgSO4), filtered and the solvent removed by means of a rotary evaporator in a water bath at a temperature of 25° C. 215 mg of organic extract was obtained.
  • This diethyl ether extract was submitted for testing.
  • the diethyl ether extract prepared directly from ground plants (WG-I-94B) was evaluated for its anti-inflammatory properties in the NF- ⁇ B Transcription assay (see protocol below). A significant inhibition activity was observed, with an estimated IC 50 of 14.3 ⁇ g/mL (see Table 15 below) in this assay in the absence of cytotoxicity at concentrations up to 100 ⁇ g/mL. Cyclosporin A was used as the reference compound (IC 50 of 0.0608 ⁇ M) (see Table 15 below) in this assay. The inhibition and cytoxicity results are shown in Table 16 and concentration response curves are shown in FIGS. 8 and 9 . Based on these results, it was established that the diethyl ether extract was effective in the inhibition of NF- ⁇ B.
  • the fresh rhizomes and roots of the plant (300 g), Siphonochilus aethiopicus , were washed with water and air dried. The freshly roots were separated from the rhizomes. The roots were crushed and the rhizomes were sliced into slices having a thickness of approximately 2-3 mm each. Both the roots and rhizomes were steam stilled in a steam distillation unit for 3 h. The crystals which formed in the condenser were washed out with deionised water and filtered through a sintered glass funnel. The structure of the crystals is given in FIG. 1 (Sample no: WG-I-94B).
  • the mixture of essential oil and water in the separating funnel condensate was allowed to stand at room temperature for 18 h.
  • the resulting crystals present on the surface, comprising the compound of formula 1 were removed by draining of the condensate and filtering using a sintered glass funnel. A total of 240 mg of the crystals was obtained. The crystals were submitted for testing.
  • the compound of formula 1 was evaluated for its anti-inflammatory properties in the NF- ⁇ B cellular transcription response assay (see Attachment I for protocol) as well as the corresponding NF- ⁇ B cellular transcription response cytotoxicity assay.
  • the compound caused >100% inhibition at 100 ⁇ M in the NF- ⁇ B cellular transcription response assay, with no apparent cytotoxicity at the same concentration. This shows that compound inhibition in the NF- ⁇ B cellular transcription is not due to general cytotoxicity.
  • An IC 50 of 15.6 ⁇ g/mL was calculated in this assay and Cyclosporin A was used as the reference compound (IC 50 of 0.0608 ⁇ M).
  • the results and concentration response curves are shown in Attachment II. Based on these results, it was established that the compound of formula 1 was effective in the inhibition of NF- ⁇ B.
  • the compound, a sesquiterpenoid, isolated from Siphonochilus aethiopicus was evaluated in in vitro assays that play a central role in chronic inflammatory diseases, such as asthma. Significant inhibition activity was observed in the NF- ⁇ B cellular transcription response assay with no cytotoxic effects.
  • the compound isolated from steam distillation of the freshly prepared roots and rhizomes
  • NF- ⁇ B represents a master regulator of inflammation and is, therefore, an attractive target for drug development.
  • cytoplasmic location of the nuclear transcription factor NF- ⁇ B is bound by an inhibitory subunit I ⁇ B; binding of I ⁇ B effectively masks the nuclear localization sequences present on the P50 and P65 subunits of NF- ⁇ B, preventing nuclear translocation.
  • I ⁇ B inhibitory subunit
  • a signal transduction pathway is activated leading to phosphorylation of key serine residues in the I ⁇ B polypeptide whereupon the NF- ⁇ B-I ⁇ B complex dissociates, I ⁇ B is rapidly degraded, and the unmasked nuclear localization signal allows NF- ⁇ B to translocate into nuclei and activate the transcription of specific genes.
  • NF- ⁇ B regulates many pro-inflammatory and pro-thrombic factors produced by activated leukocytes.
  • NF- ⁇ B represents a master regulator of inflammation and is, therefore, an attractive target for drug development.
  • Test compound and/or vehicle are incubated with the cells (1.5 ⁇ 10 6 /ml) in the presence of 0.5 ⁇ M A23187 and 50 ng/ml PMA (phorbol 12-myristate 13-acetate) in RPMI-1640 pH 7.4 at 37° C. for 4 hours.
  • Test compound-induced ⁇ -galactosidase activity is determined by the conversion of FDG (fluorescein di- ⁇ -D-galactopyranoside) to fluorescein.
  • Fluorescence intensity is read on SpectroFluor Plus plate reader. Decrease of 50 percent or more (>50%) in fluorescence intensity, relative to 10 ⁇ M cyclosporin A, indicates significant inhibitory activity. Compounds are screened at 10, 1, 0.1, 0.01 and 0.001 ⁇ M. These same concentrations are concurrently applied to a separate group of treated cells and evaluated for possible compound-induced cytotoxicity only if significant stimulation or inhibition is observed (Cat. #361100).
  • cytokines exist that are known mediators of inflammation and are involved in the asthmatic disease process.
  • a causative agent like an allergen
  • the bronchial epithelial cells are activated and produce certain pro-inflammatory cytokines (interleukins, abbreviated IL) in particular the chemokine IL-8.
  • IL-8 pro-inflammatory cytokines
  • WG-I-101 diethyl ether extract
  • WG-I-101 was suspended in 0.5% CMC/0.1% Tween 80.
  • the test substance at a dose of 500 mg/kg was given orally twice daily for 3 consecutive days and an additional final dose was added at one hour before challenge of Ovalbuminon testing day.
  • Dosing volume of 10 ml/kg was used. The formulation is summarized as follows:
  • Test Light Formula Compound Vehicle Solubility (a) Colour Protection (b) Temperature (c) mg/ml WG-I-101 0.5% I Brown Y RT 50 CMC/0.1% Tween 80 (a) This is based upon visual observation S: soluble; I: Insoluble ppt: precipitation (b) Y: formula is kept in tube or vial with brown colour, or covered with aluminium foil (c) TR: room temperature; prepared and stored under 15° C. ⁇ 30° C. Temperature throughout experiment, lower or higher than room temperature is specified.
  • a carotid artery was cannulated (PE50, Clay Adams, USA) for measurements of blood pressure using a Stathem P23 ⁇ L transducer (Viggo-Spectramed, USA) and heart rate was obtained from lead 11 ECG.
  • Guinea pigs were sensitized with IP injections of ovalbumin (0.5 ⁇ g/0.5 ml/animal) and Al(OH) 3 (1 mg/0.5 ml/animal) on days 1 and 8. The animals were then challenged with 15 ⁇ g/kg of Ovalbumin (IV) between days 19 and 23 and bronchopulmonary constriction was recorded as an increase in ITP.
  • test substance at 500 mg/kg was administered orally twice daily for 3 consecutive days and an additional final dose was added on testing day, then, one hour after final dose, the animals were injected intravenously (1 ml/kg) with a “cocktail”: Indomethacin (10 mg/kg), Mepyramine (2 mg/kg), and Propranolol (100 ⁇ g/kg), followed by challenge with ovalbumin 5 minutes later.
  • a “cocktail” Indomethacin (10 mg/kg), Mepyramine (2 mg/kg), and Propranolol (100 ⁇ g/kg)
  • challenge resulted in bronchoconstrictor responses (increase in ITP) ranging from 45 to 85 percent of maximum possible bronchoconstriction as measured by complete tracheal occlusion.
  • a 50 percent or more ( ⁇ 50%) inhibition of the ovalbumin-induced bronchoconstriction relative to the vehicle-treated control animals is considered significant.
  • Bronchoalveolar lavage fluid was retrieved after instilling 5 ml of phosphate buffered saline for 2 times.
  • TNF- ⁇ , IL-1 and IL-8 in bronchoalveolar lavage fluid (BALF) supernatants were measured with ELISA after ovalbumin challenge.
  • One-way ANOVA followed by Dunnett's Test was applied for comparison between the vehicle control and test compound-treated groups. P ⁇ 0.05 is considered significant.
  • Anesthetized and artificially ventilated guinea pigs previously sensitized were pretreated with Mepyramine 2 mg/kg, Indomethacin 10 mg/kg and Propranol 0.1 mg/kg for 5 minutes; arterial blood pressure (BP, mm Hg), heart rate (HR, beats/min.) and tracheal pressure (TP, cm H2O) were recorded after oral dosing for 3 days.
  • BP arterial blood pressure
  • HR heart rate
  • TP tracheal pressure
  • Percent (%) inhibition is calculated according to the formula of [Increase in Tracheal Pressure (vehicle-treated)] ⁇ [Increase in Tracheal Pressure (test substance-treated)/[Increase in Tracheal Pressure (vehicle-treated)] ⁇ 100%.
  • Bronchoalveolar lavage fluid (BALF) samples in the WG-I-101-treated group were prepared for measurements of TNF- ⁇ , IL-1 ⁇ and IL-8 after ovalbumin challenge.
  • One-way ANOVA followed by Dunnett's test was applied for comparison between the vehicle control and test compound-treated groups. P ⁇ 0.05 is considered significant.
  • WG-I-101 caused great inhibition of IL-8 production (P ⁇ 0.05) versus control after ovulbumin challenge in guinea pigs, without significant effects on TNF- ⁇ and IL-1 ⁇ secretion. Results are shown in Table 19.
  • TNF- ⁇ , IL-1 ⁇ and IL-8 levels of each sample were assessed using TNF- ⁇ , IL-1 ⁇ and IL-8 ELISA kits, respectively.
  • One way ANOVA followed by Dunnett's Test was applied for comparison between the vehicle control and test compound-treated groups. P ⁇ 0.05 is considered significant.
  • Groups of 5 male Dunkin-Hartley guinea pigs weighing 250 ⁇ 50 g are anesthetized with pentobarbital sodium (50 mg/kg i.p., plus an additional 15 mg/kg i.p. if required) and succinylcholine chloride (2 mg/animal i.p.) is subsequently administered to prevent spontaneous respiration.
  • Body temperature is maintained at 370 to 38° C.
  • the trachea is cannulated and the guinea pig ventilated with a Harvard rodent respirator in a closed system. Tracheal pressure is recorded through a side-arm of the cannula connected to a P23ID Statham transducer. Respiratory rate set at 50 strokes/minute with a stroke volume (approximately 1 ml/100 g) sufficient to produce a baseline tracheal pressure of 6 cm H 2 O. Mean arterial pressure is monitored from a cannulated carotid artery, and heart rate is obtained from chest electrodes arranged for lead II. The jugular vein is cannulated for i.v. vehicle or drug administration in a volume of 1 ml/kg.
  • Lipoxygenase is activated and resultant leukotrienes generated through antigen challenge in previously sensitized animals.
  • Guinea pigs are sensitized with injections of ovalbumin and Al(OH) 3 (0.5 ⁇ g and 1 mg, respectively in a volume of 0.5 ml/animal i.p.) on day 1, boosted with the same dose of ovalbumin and Al(OH) 3 on day 8, and challenged between day 19 and 23 with ovalbumin (50 ⁇ g/kg i.v.) to effect a maximal-induced airway constriction, reflected as an increase in tracheal pressure (cm H 2 O). Also, an increase in tracheal pressure could be related to an increase in stiffness of the respiratory system.
  • the animals are pretreated with CSIR compound by oral gavage for two days. On the day of experiment, the animals are administered with CSIR compound 30 min before anesthesia and surgery which takes 15 min. The ovalbumin challenge is administered 60 min after dosing with CSIR.
  • the animals are pretreated 5 minutes before test substance administration with i.v. indomethacin (10 mg/kg), mepyramine (2 mg/kg), and propranolol (0.1 mg/kg): a “cocktail” designed to inhibit the generation of cyclooxygenase products (thromboxanes, etc.) as well as antagonize histamine and ⁇ -adrenergic receptors.
  • antigen challenge results in airway constrictor responses ranging form 45 to 85 percent of maximum possible airway constriction obtained by tracheal occlusion.
  • Phenidone the positive standard, is administered i.v. (30 mg/kg) 5 minutes before ovalbumin challenge in 5 guinea pigs. A 50 percent or more ( ⁇ 50) inhibition of the induced airway constriction relative to vehicle treated control animals is considered significant.
  • Test substances active in this model system indicate possible in vivo inhibition of lipoxygenase activity and/or antagonism of its leukotriene(s) generated receptor activation. Consequently, it may be worthwhile to compare results in this model system with those obtained from: cyclooxygenase inhibition to determine specificity for enzyme inhibition; leukotriene D 4 antagonism to determine test substance action on enzyme end-product receptor activation relative to lipoxygenase enzyme inhibition; and anticholinergic activity and/or antihistamine H 1 and H 3 activity to determine selectivity for receptor antagonism and/or bronchodilator action.
  • the extracts of the invention are non-steroidal components isolated in a natural form, but having the same mode of action as steroidal compounds commonly used for the preventative treatment of allergic disease, such as asthma and atopy.
  • sesquiterpenoid compound of formula 1 of the invention shows activity as a leukotriene modifier, inhibiting the actions of inflammatory mediators and leukotrienes.
  • the extract When used in the form of an essential oil, the extract has a combined effect, in that reduction of asthma and allergy inflammation occurs through bronchodilation, inhibition of leukotriene biosynthesis, as well as due to downregulation of glucocorticoid receptors.
  • a diethyl ether extract containing the sesquiterpenoid compound of formula 1 of the invention shows significant inhibition activity in the NF- ⁇ B Transcription Response cellular assay with no cytotoxic effects.
  • the diethyl ether extract can thus be used to inhibit specific activity of the NF- ⁇ B Transcription Response, thereby inhibiting the release of various pro-inflammatory and inflammatory mediators such as the cytokine Interleukin-8 (IL-8) that are responsible for the inflammatory pathway of asthma.
  • IL-8 cytokine Interleukin-8
  • a diethyl ether extract containing the sesquiterpenoid compound of formula 1 of the invention shows significant inhibition in the production of IL-8 an inflammatory cytokine responsible for the inflammatory pathway of asthma.
  • the invention may be effective in the overall preventative treatment and remission of allergic diseases such as asthma, adding a useful tool in the management of these diseases.
  • Remission of allergic diseases and asthma is specifically defined for the purposes of this specification as a symptom-free period without subjects taking conventionally available treatment. This remission period is longer than the period that is obtained using conventional treatment for the management of allergic diseases such as asthma.
  • the invention in the form of organic solvent extracts, dried rhizomes, an essential oil, or as the single chemical compound of formula 1, either used alone or in combination, or as compositions, provides an alternative to the use of steroidal anti-inflammatory drugs such as glucocorticoids for the long-term preventative treatment and remission of allergic diseases, especially asthma and atopy.
  • steroidal anti-inflammatory drugs such as glucocorticoids
  • glucocorticoid receptor level typically are of a steroidal nature and have numerous detrimental side effects during long-term use thereof.
  • the compounds, derivatives, compositions and products of the invention are non-steroidal in nature, yet, surprisingly, have been found to act efficiently at the glucocorticoid receptor (GR) level.
  • GR glucocorticoid receptor

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pulmonology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Immunology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)
US12/281,517 2006-03-03 2007-02-28 Preventative treatment and remission of allergic diseases Abandoned US20090082433A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
ZA200601860 2006-03-03
ZA2006/01860 2006-03-03
PCT/IB2007/050649 WO2007113698A2 (en) 2006-03-03 2007-02-28 Preventative treatment and remission of allergic diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2007/050649 A-371-Of-International WO2007113698A2 (en) 2006-03-03 2007-02-28 Preventative treatment and remission of allergic diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/718,190 Division US8158165B2 (en) 2006-03-03 2010-03-05 Preventative treatment and remission of allergic diseases

Publications (1)

Publication Number Publication Date
US20090082433A1 true US20090082433A1 (en) 2009-03-26

Family

ID=38521466

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/281,517 Abandoned US20090082433A1 (en) 2006-03-03 2007-02-28 Preventative treatment and remission of allergic diseases
US12/718,190 Active 2027-08-26 US8158165B2 (en) 2006-03-03 2010-03-05 Preventative treatment and remission of allergic diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/718,190 Active 2027-08-26 US8158165B2 (en) 2006-03-03 2010-03-05 Preventative treatment and remission of allergic diseases

Country Status (8)

Country Link
US (2) US20090082433A1 (pt)
EP (1) EP2007408B1 (pt)
CN (1) CN101432009B (pt)
AP (1) AP2893A (pt)
BR (1) BRPI0708554B8 (pt)
RU (1) RU2008137281A (pt)
WO (1) WO2007113698A2 (pt)
ZA (1) ZA200807874B (pt)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013150406A2 (en) 2012-04-04 2013-10-10 Integral Bioceuticals (Pty) Ltd Siphonochilone and related compounds and uses thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2495058B (en) 2010-07-26 2014-03-05 Seven Networks Inc Context aware traffic management for resource conservation in a wireless network
WO2013015835A1 (en) 2011-07-22 2013-01-31 Seven Networks, Inc. Mobile application traffic optimization
EP2599003B1 (en) 2010-07-26 2018-07-11 Seven Networks, LLC Mobile network traffic coordination across multiple applications
CN108400946B (zh) 2013-06-11 2019-09-17 杭州硕文软件有限公司 一种用于减少网络通信量的方法、装置、系统及介质

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09124677A (ja) * 1995-11-01 1997-05-13 Eba Keshohin:Kk ゲットウを用いた組成物及びその製造方法
JPH09143086A (ja) * 1995-11-22 1997-06-03 San Eng:Kk 皮膚疾患治療剤及び化粧料
JP2002512198A (ja) * 1998-04-16 2002-04-23 アイダ ディベロップメント アクティーゼルスカブ アルピニアガランガ中に存在する芳香族化合物及びテルペノイドを含む新規相乗組成物
WO2001030367A1 (fr) * 1999-10-27 2001-05-03 Shiseido Company, Ltd. Antagonistes du facteur d'activation des plaquettes et compositions dermatologiques destinees a un usage externe contenant lesdits antagonistes
JP2001187725A (ja) * 2000-01-06 2001-07-10 Pola Chem Ind Inc ストレス予防剤及びそれを含有してなる皮膚外用剤
JP2001192338A (ja) * 2000-01-11 2001-07-17 Pola Chem Ind Inc ストレスの悪影響からの回復促進剤及びそれを含有してなる皮膚外用剤
JP2001261545A (ja) * 2000-03-21 2001-09-26 Shiseido Co Ltd 肌荒れ防止用皮膚外用剤
JP2003111581A (ja) * 2001-10-02 2003-04-15 Saburo Toyoda 健康飲料
JP2004269450A (ja) * 2003-03-11 2004-09-30 Shiseido Co Ltd 新規化合物およびケモカイン発現阻害剤
JP4495406B2 (ja) * 2003-04-21 2010-07-07 株式会社武蔵野免疫研究所 センダングサ属植物抽出物含有組成物
JP4073358B2 (ja) * 2003-04-28 2008-04-09 株式会社武蔵野免疫研究所 化粧品

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013150406A2 (en) 2012-04-04 2013-10-10 Integral Bioceuticals (Pty) Ltd Siphonochilone and related compounds and uses thereof

Also Published As

Publication number Publication date
WO2007113698A3 (en) 2008-01-03
WO2007113698A2 (en) 2007-10-11
CN101432009B (zh) 2012-03-21
RU2008137281A (ru) 2010-04-10
EP2007408B1 (en) 2019-05-01
BRPI0708554A2 (pt) 2011-05-31
BRPI0708554B1 (pt) 2021-01-19
AP2008004630A0 (en) 2008-10-31
US8158165B2 (en) 2012-04-17
EP2007408A2 (en) 2008-12-31
ZA200807874B (en) 2009-04-29
BRPI0708554B8 (pt) 2021-05-25
CN101432009A (zh) 2009-05-13
AP2893A (en) 2014-05-31
US20100168227A1 (en) 2010-07-01

Similar Documents

Publication Publication Date Title
US8158165B2 (en) Preventative treatment and remission of allergic diseases
Shirole et al. Investigation into the mechanism of action of essential oil of Pistacia integerrima for its antiasthmatic activity
Tong et al. Therapeutic effects of Caesalpinia minax Hance on complete Freund's adjuvant (CFA)-induced arthritis and the anti-inflammatory activity of cassane diterpenes as main active components
TWI444194B (zh) 抗癌萃取物及化合物
BRPI0608546A2 (pt) composição farmacêutica, e, uso de isoorientina derivada naturalmente
JP5478486B2 (ja) 植物抽出物及びその治療的使用
US20110117121A1 (en) Compositions for treatment and inhibition of pain
CN105531282A (zh) 治疗与线粒体功能或抑制PDE4、PDE5和IKK-β相关的病况的源自茶藨子属物种的生物碱组合物
AU2014351474A1 (en) Salacia compositions, methods of treatment by their administration, and methods of their preparation
WO2008125928A2 (en) Standardized bioactive extracts of annona squamosa
KR20140102599A (ko) 뇌암 치료를 위한 방법 및 조성물
EP1981517B1 (en) Herbal composition
JP2000154151A (ja) 免疫抑制剤
CN102000073A (zh) 黄连碱在预防和治疗心肌缺血性疾病中的应用
CN103494806A (zh) 苯骈α-吡喃酮类化合物的应用及其制备方法
KR20230088290A (ko) 사철쑥 및 죽순 추출물을 포함하는 장질환 예방 또는 치료용 조성물
CN114728030B (zh) 用于预防、缓解或治疗呼吸系统疾病的组合物
KR100361090B1 (ko) 꾸지나무로부터 분리한 항염증 활성을 나타내는 신규한플레닐레이티드 플라보노이드 화합물 및 이를 주성분으로함유하는 꾸지나무 추출물, 이들의 제조방법 및 이들을함유하는 약학적 조성물
JP2005501079A (ja) 気管支呼吸困難の治療及び療法のための草本組成物
Yingzhan et al. Total glucosides of Rhizoma Smilacis Glabrae: a therapeutic approach for psoriasis by regulating Th17/Treg balance
KR20150002685A (ko) 동맥경화성 혈관 질환을 치료하기 위한 방법 및 조성물
Rafi et al. Acmella calva (DC.) RK Jansen Flower: A Comprehensive In vivo Study of its Central and Peripheral Analgesic, Hypoglycemic, Antidepressant, and Anti-Diarrheal Properties
KR101533487B1 (ko) 개가시나무 추출물 또는 이로부터 분리된 폴리페놀성 화합물을 유효성분으로 포함하는 항염증 및 항요로결석 조성물
CN110123797B (zh) 二氢查尔酮类化合物Renifolin F的新用途
WO2005084693A1 (fr) Extrait de la graine de hippophae rhamnoides

Legal Events

Date Code Title Description
AS Assignment

Owner name: CSIR, SOUTH AFRICA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HORAK, ROELOF MARTHINUS;WADIWALA, EBRAHIM;FOUCHE, GERDA;AND OTHERS;REEL/FRAME:021988/0667

Effective date: 20081208

STCB Information on status: application discontinuation

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