EP2254899A1 - Blocking the metastasis of cancer cells and the uses of new compounds thereof - Google Patents

Blocking the metastasis of cancer cells and the uses of new compounds thereof

Info

Publication number
EP2254899A1
EP2254899A1 EP09721583A EP09721583A EP2254899A1 EP 2254899 A1 EP2254899 A1 EP 2254899A1 EP 09721583 A EP09721583 A EP 09721583A EP 09721583 A EP09721583 A EP 09721583A EP 2254899 A1 EP2254899 A1 EP 2254899A1
Authority
EP
European Patent Office
Prior art keywords
cancer
angeloyl
alkanoyl
benzoyl
angiopoietin
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.)
Withdrawn
Application number
EP09721583A
Other languages
German (de)
French (fr)
Other versions
EP2254899A4 (en
Inventor
Pui-Kwong Chan
May Sung MAK
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.)
Pacific Arrow Ltd
Original Assignee
Pacific Arrow Ltd
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
Priority claimed from PCT/US2008/002086 external-priority patent/WO2008133766A1/en
Priority claimed from US12/344,682 external-priority patent/US8841265B2/en
Application filed by Pacific Arrow Ltd filed Critical Pacific Arrow Ltd
Publication of EP2254899A1 publication Critical patent/EP2254899A1/en
Publication of EP2254899A4 publication Critical patent/EP2254899A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/24Condensed ring systems having three or more rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention provides methods and compositions for reducing the adhesion protein in cell and blocks the migration, metastasis of cancer cells or inhibits the growth of cancers or anti-angiogenesis; wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers, wherein the adhesion proteins or receptors comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • the composition from Maesa balansae for anti-leishmanial is a United National funding project.
  • This invention is providing methods for inhibiting cancer metastasis or growth, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers, wherein the method is using the extract, composition and compounds from Maesa balansae, Barringtonia acutangula, Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum.
  • This invention is providing methods of using the extract, composition and compounds from Maesa balansae and Barringtonia acutangula, Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum for reducing adhesion protein, for inhibiting cancer metastasis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers.
  • This invention provides a method for modulating the adhesion of cancer cell and blocking their migration, metastasis or inhibiting the growth of cancers or anti- angiogenesis, wherein the adhesion protein and their receptors comprise fibronectin, integhns family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • This invention provides a method of antiprotozoal comprising treating leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis and malaria.
  • Figure 1 Analysis of genesis of blood vessel in xenograft tumor treated with compound Y.
  • Figure A and B show the tumor sections taking from mice without Xanifolia Y treatment.
  • Figure C and B show the tumor sections taking from mice with Xanifolia Y treatment. More blood vessels were observed in the control Groupi than those in the drug-treated Group 2
  • Figure 2 Experiment shows that Y10 is cytotoxic to Leishmania Major (promastigotes) with IC50 approximately equal to 15ug/ml.
  • Figure 3 Shows the inhibition activity of compound Y, X, ACH-Y and AKOH-Y
  • This invention provides methods and compositions for modulating the gene expression to cure diseases or reduce the syndrome of diseases, wherein the modulating comprises positive and negative regulating.
  • the method comprises inhibiting the gene expression.
  • the method comprises stimulating the gene expression.
  • This invention provides methods and compositions for inhibiting the migration, metastasis or growth of cancers or anti-angiogenesis, wherein the methods comprise affecting the gene expression, wherein comprise affecting the adhesion proteins or their receptors, reducing adhesion protien, or inhibiting the expression or secretion of adhesion proteins, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • This invention provides methods and compositions for inhibiting the migration, metastasis or growth of cancers or anti-angiogenesis, wherein the methods comprise affecting the gene expression, wherein comprises stimulating the gene expression.
  • This invention provides a method for altering the characteristic of cancer cell membrane resulting in blocking the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the method comprises reducing adhesion protiens or their receptors, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • This invention provides methods, processes, compounds and compositions of reducing adhesion protein of cells, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD54, CAM, elastin and FAK.
  • methods comprise inhibiting the gene expression.
  • this invention provides a method of reducing the secretion of fibronectin.
  • the method can block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer.
  • cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer.
  • the method is contracting the said cell with compound selected from Mb1 , Mb2, Mb2.1 , Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13, ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba17, Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof and Compound A to X and A1 to X1 in the application.
  • compound selected from Mb1 , Mb2,
  • This invention provides a method of altering the characteristic of cancer cell membrane, wherein the method comprises altering the secretion of adhesion proteins, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • the methods, processes, compounds and compositions comprises blocking, suppressing or inhibiting the expression or secretion of adhesion protein, wherein the adhesion proteins .
  • the methods, processes, compounds and compositions is interacting with adhesion protein, wherein the adhesion proteins.
  • the methods, processes, compounds or compositions can block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer.
  • the method is contracting the said cell with compound selected from Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13,, ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba 17, Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof and Compound A to X and A1 to X1 in the application.
  • compound selected from Mb1 , Mb2, Mb3, Mb4, Mb
  • the adhesion proteins help cancer cell adhesion, invasion or metastasis, wherein the cancers comprise ovarian cancer. Reducing the adhesion proteins will reduces the metastasis of cancers.
  • the fibronectin is one of the key factors in the biology of epithelial ovarian cancers. The reducing of fibronectin will inhibit the metastasis of cancer cells.
  • This invention provides a method and composition for inhibiting the secretion of adhesion protein comprising fibronectin in order to cure the diseases, wherein the diseases comprise inhibiting cancer growth, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer.
  • the method is contracting the said cell with compound selected from Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, MbI O 1 Mb1 1 , Mb12 and Mb13, ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5, ACH-MbI 3, Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba 17, Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof and Compound A to X and A1 to X1 in the application.
  • compound selected from Mb1
  • This invention provides a use of compound for manufacture of medicament or a composition for inhibiting the growth, migration, metastasis of cancer and by altering the characteristics of membrane of cancer cell, wherein the characteristics comprise adhesion of proteins; wherein comprising the secretion of proteins or the adhesion of cells; wherein the characteristic comprise adhesion ability; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK; wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer; wherein the method is administering contacting the say
  • the present invention provides vaccine for cancer immunotherapy.
  • the vaccines comprise extract, compositions, compounds and saponins from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula.
  • the compounds can be obtained from synthesis, semi-synthesis or modification.
  • the method comprises administering to the said subject and effective amount of vaccine for enhancing the immune response.
  • the vaccines comprise saponins isolated from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula.
  • the present invention provides adjuvant compositions for cancer curing.
  • the adjuvant compositions comprise extract, compositions, compounds and saponins from
  • the method comprises administering to the said subject and effective amount of the above adjuvant compositions for enhancing the immune response.
  • vaccine compositions comprise inhibiting cancer metastasis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers.
  • the present invention provide a use of compound for manufacture of medicament or methods for making vaccine, wherein the vaccine comprise compounds or saponins from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula, wherein the vaccine can be used for inhibiting cancer growth, wherein the vaccine is having immune adjuvant activity, wherein the saponins comprise Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof.
  • the saponin may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound(s) are selected from Compound Z1 to Z13 in the application.
  • the saponin comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13,.
  • the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety.
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the compound(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5, and ACH-MbI 2.
  • the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16 and Ba 17.
  • the compound(s) are selected from Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof.
  • this invention provides a method for antiparasitics by using the above compounds, wherein the antiparasitics comprise inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria; wherein a method comprises contacting cells with an effective amount of an isolated, purified or synthesized compound, or its salt, or ester thereof, selected from the above compounds.
  • This invention provides a use of compound for manufacture of medicament or a method for pharmaceutical composition useful for inducing an immune response to and antigen in an individual comprising the saponin composition from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae or Barringtonia acutangula or synthesis.
  • the present invention provide methods for enhancing an immune response to an antigen in an individual comprising administering an effective amount of saponins/compositions from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula, or the saponin comprise Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof, or the saponin may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound(s) are selected from Compound Z1 to Z13 in the application.
  • the saponin comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13,
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the composition(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5, and ACH-MbI 2.
  • the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba17.
  • This invention is providing methods or a use of compound for manufacture of medicament for modulating adhesion or angiogenesis of cancer cells, antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, inhibiting cancer metastasis or growth, of using the extract, composition and compounds from Maesa balansae, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers;
  • the method is modulating the phosphatidylinositol signaling system, regulate the gene expression of RGS4 , LEPR, ICFBP3, ANGPT2, GPNMB, NUPR1 or LOC100126784.
  • the compounds can be purified from natural resource comprising Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae or Barringtonia acutangula, or synthesized.
  • the compounds comprise following: (Our purification methods and biological assays including the MTT assay in International Application No. PCT/US05/31900, filed September 7, 2005, U.S. Serial No. 1 1 /289142, filed November 28, 2005, and U.S. Serial No. 1 1/131551 , filed May 17, 2005, and PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008, the contents of which are incorporated herein by reference)
  • R3 is OH, R4 is OH, also named Mb1
  • R3 is OH, R4 is OH, also named Mb2
  • R3 is
  • R1 is OH
  • R2 is O-benzoyl
  • R3 is O-benzoyl
  • R1 is O-acetyl
  • R2 is O-benzoyl
  • R3 is O-benzoyl 3-O-[ ⁇ -D-galactopyranosyl(1 ⁇ 2)]- ⁇ - D-xylopyranosyl (1 ⁇ 3)- ⁇ -D-glucuronopyranosyl-
  • R1 is OH
  • R2 is O-benzoyl
  • R3 is O-benzoyl
  • R1 is O-acetyl
  • R2 is O-benzoyl
  • R3 is O-benzoyl
  • the anti-cancer activity of Mb's compounds with ES2 cell the IC50 of Mb1 is 8ug/ml, Mb2 is /ml, Mb3 is 8ug/ml, Mb4 is 15ug/ml, Mb5 is 6.5ug/ml, and Mb6 is 10ug/ml, Mb7 is 12ug/ml, Mb8 is 20ug/ml, Mb9 is 18ug/ml, Mb12 is 10ug/ml.
  • the anti-cancer activity of ACH's compounds with ES2 cell the IC50 of ACH-Z4 is 40 ug/ml, ACH-Y3 is 20 ug/ml, ACH-Y10 is 20 ug/ml, ACH-Y2 is 35 ug/ml, ACH-Y8 is 35 ug/ml, ACH-Y7 is 65 ug/ml, ACH-YO is 20 ug/ml, ACH-X is 40ug/ml, ACH-E is 60 ug/ml
  • Ba1 is 5 ug/ml
  • Ba2 is 5 ug/ml
  • Ba3 8 ug/ml
  • Ba 5 is 16 ug/ml
  • Ba7 is 1 1 ug/ml
  • Ba8 is 20 ug/ml
  • Ba9 12 ug/ml
  • Ba17 5 ug/ml.
  • the anti-cancer activity of Xanifolia Y with cells the IC50 of Y on (bladder)TB9 cells is 5 ug/ml; IC50 of Y on (lung)H460 cells is 7.5 ug/ml; IC50 of Y on HeLa cells is 20 ug/ml; IC50 of Y on skin cells is 12 ug/ml; IC50 of Y on ES2(ovarian) cells is 5 ug/ml; IC50 of pure Y on (Mouth) KB cells is 6 ug/ml; IC50 of Z12 on ES2(ovarian) cells is 16 ug/ml; Z4 is 20ug/ml
  • IC50 of Mb5 (bladder)TB9 is 6.5ug/ml, (Prostate) DU145 is 7.6 ug/ml, (Lung) H460 is 12 ug/ml (Liver) HepG2 is 6.5 ug/ml, (brain) T98G is 12 ug/ml, (Skin) SK-MELS is 25 ug/ml, (Ovary)ES2 is 6.5 ug/ml, (Breast)MCF7 is 1 1 ug/ml.
  • IC 50 of ACH-Mb5 (bladder)TB9 is 5.7ug/ml, (Prostate) DU145 is 6.4 ug/ml, (Lung) H460 is 6.5 ug/ml (Liver) HepG2 is 4 ug/ml, (brain) T98G is 6 ug/ml, (Skin) SK-MELS is 22 ug/ml, (Ovary)ES2 is 8 ug/ml, (Breast) M C F7 is 13 ug/ml.
  • This invention provides a method and composition for reducing of adhesion protein to cure the diseases, wherein the diseases comprise inhibiting cancer growth, reducing leg swelling, symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, for reducing symptoms of pain; thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, comprising administering to a subject, in need thereof, an effective amount of the composition of this invention.
  • the method comprises interacting with adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • adhesion protien comprising fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • this invention provides a method of reducing the secretion of fibronectin.
  • the method comprises reducing the adhesion ability of adhesion protein; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • the method comprises modulating the secretion of adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • the method comprises blocking the secretion of adhesion protien, wherein the adhesion protein comprising fibronectin.
  • the method is administering contacting an effective amount of the compound selected from formulas in this application.
  • this invention provides a method and composition for modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition.
  • this invention provides a method of antiprotozoal comprising treating leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis and malaria.
  • This invention provides a use of compound for manufacture of medicament, a method and composition for altering the characteristic of adhesion protein to cure diseases, wherein the characteristic comprising adhesion ability, wherein the method comprises reducing the secretion of fibronectin, wherein the diseases comprise inhibiting cancer growth, reducing leg swelling, symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, for reducing symptoms of pain; thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, comprising administering to a subject, in need thereof,
  • a process and method for administration of the composition wherein administration is by intravenous injection, intravenous drip, intraperitoneal injection or oral administration; wherein administration is by intravenous drip- 0.05-0 2mg/kg compound dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution, or by intravenous injection: 0.05-0.2mg/kg/day compound dissolved in 10-2OmI of 10% glucose solution or of 0.9% NaCI solution, or by intravenous drip: 0.1 -0.2mg/kg/day compound dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution, or by intravenous injection: 0.1 -0.2mg/kg/day compound dissolved in 1 0-2OmI of 10% glucose solution or of 0 9% NaCI solution, or by intraperitoneal injection(I.P ): 2 5mg/kg/day compound dissolved in 10% glucose solution or of 0.9% NaCI solution, or by oral administration wherein the dosage of mammal is 1 -10mg/Kg
  • R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O- alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O- alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof;
  • R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl,
  • R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof; in embodiment, the acyl has carbon 2 to 10.
  • the method is administering contacting the compounds, wherein the compound is selected from the following: a) An isolated, purified or synthesized compound is having structure Xanifolia(Y),
  • the method is administering contacting the compound, wherein the compound is selected from the following: k) An isolated, purified or synthesized compound is having a structure:
  • the method is administering contacting the compound, wherein the compound is isolated, purified or synthesized having a structure selected from following formulas: wherein R1 , R2 are individually selected of an O-acetyl or O-angeloyl; wherein the R3,
  • R4, R5, R6, R7 is hydrogen or hydroxyl
  • the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof.
  • the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety.
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the compound(s) are selected from Compound Z1 to Z13 in the application.
  • the method is administering contacting the compound comprise of a triterpene wherein the carbon position 21 , 21 has an unsaturated group and sugar moieties at carbon 3.
  • methods and compounds of this application reducing the adhesion ability inhibit bacteria in colonization and regulate tropism of cells.
  • This invention provides a use of compound for manufacture of medicament, a method and composition for modulating adhesion or anti-angiogenesis of cancer tumor, antiparasitics or manufacturing an adjuvant composition, wherein the modulating adhesion of cancer cell comprising modulating the secretion or expression of adhesion protein, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, cadherin, heparin, tenascin, CD 54, CAM; wherein the modulating comprises reducing, inhibiting and stimulating, wherein modulating adhesion protein comprises reducing the fibronectin for inhibiting the metastasis or growth of cancer cells, wherein the cancer is selected from breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, and renal cancer; wherein the modulating adhesion of cancer cell comprising
  • the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety.
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the compound(s) are selected from Compound Z1 to Z13 in the application.
  • the compound(s) are selected from ACH-Z4, ACH- Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5 and ACHMbI 2.
  • the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, and Ba17.
  • the saponins comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12, and Mb13.
  • the methods and compositions of this application can be used for manufacturing an adjuvant vaccine, wherein the methods and compositions are used for manufacturing an adjuvant vaccine in a subject, wherein the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof.
  • the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety.
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the compound(s) are selected from Compound Z1 to Z13 in the application.
  • the compound(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5 and ACH-MbI 2.
  • the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, and Ba 17.
  • the saponins comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13.
  • this application provides an adjuvant composition comprising a saponin or compound selecting from above; wherein the composition is comprised of an immunostimulatory oligonucleotide.
  • the methods and compositions of this application can be used for manufacture of medicament for vaccine or antiviral agent for Enterovirus comprising EV71 , wherein the method is administering contacting the cell or subject with compounds selected in this application comprising Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13.
  • the composition comprises the bioactive compounds from natural plants or synthesis.
  • the majority of the plants are from the Sapindaceae family, which has 140-150 genera with 1400-2000 species.
  • the program is based on our purification methods and biological assays including the MTT assay See International Application No. PCT/US05/31900, filed September 7, 2005, U.S. Serial No. 1 1 /289142, filed November 28, 2005, and U.S. Serial No. 1 1 /131551 , filed May 17, 2005, and PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008, the contents of which are incorporated herein by reference.
  • This invention provides a composition comprising an effective amount of triterpenoidal saponins named as Xanifolia Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, and YO or their salt or their derivatives.
  • the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, and Ba17 or their salt or their derivatives.
  • the saponins comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13, or their salt or their derivatives for modulating the adhesion protein, reducing adhesion protein or reducing the secretion of fibronectin, for treating chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrheal, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of pain; for reducing symptoms of stomach pain; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, thrombosis, thromophlebitis; for treating
  • the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof.
  • the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety.
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the compound(s) are selected from ACH-Z4, ACH- Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5 and ACH-MbI 2.
  • the compounds of this invention can be isolated from natural sources or synthesized.
  • the saponin will be partial hydrolysis to a mixture of products.
  • the procucts can be separated by HPLC.
  • specific partial hydrolysis can be achieved with enzymes.
  • the ⁇ -glucosidase is good enzyme to cleave of the ⁇ -glucose from saponin.
  • This invention provides a use of compound for manufacture of medicament or a method of modulating the adhesion proteins or their receptors, reducing the adhesion ability of the cancer cells, wherein the modulating comprises the positive or negative regulating.
  • the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • the method is reducing the secretion of fibronectin.
  • This invention provides a method of blocking the migration, metastasis of cancer cells or inhibiting cancer cell growth or inhibiting leishmaniases modulating adhesion or inhibiting angiogenesis of cancer tumor, antiparasitics or manufacturing an adjuvant composition comprising administering an effective amount of a pharmaceutical composition comprising a composition comprises the molecular formula or compound in this invention.
  • the cancers comprise Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma cancer, ovarian cancer, renal cancer, Prostate cancer, Breast cancer, bladder cancer, cervix cancer, liver cancer, bone cancer, brain cancer and Skin cancer.
  • the compounds of this invention can be isolated from natural sources or synthesized.
  • the method is administering contacting the compounds, wherein the compound is selected from the following:
  • This invention provides uses of a compound for manufacture of medicament selected from formula (1 B), for modulating, regulating or interact with the adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • this invention provides a method of reducing the secretion of fibronectin, modulating adhesion or angiogenesis of cancer cells, antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, inhibiting cancer metastasis or growth, using the compounds selected for the following:
  • R1 comprises a group selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, dibenzoyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, acyl, aryl, heterocylic, heteroraryl, alkenylcarbonyl and derivatives thereof
  • R2 comprises a group selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkenylcarbonyl and derivative thereof
  • R4 represents CH 2 OR6 or COOR
  • R1 comprises a sugar moiety wherein substituted with two groups selecting from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic heteroraryl and a derivative thereof.
  • R1 comprises a sugar moiety wherein substituted with at least one group selecting from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R2 comprises a sugar moiety wherein at least one group is selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R2 comprises a sugar moiety or a side chain wherein at least two groups are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R4 comprises CH 2 OR6 or COOR6 wherein R6 is a sugar moiety which comprises at least one group selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R6 is a sugar moiety which comprises at least one group selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R4 comprises CH 2 OR6 orCOOR6, wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substitutedalkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R4 comprises CH 2 OR6 or COOR6, wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl and senecioyl.
  • R4 comprises CH 2 OR6 or COOR6 of formula (1 B), at least two of R1 , R2 and R6 comprise the group selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof.
  • R4 comprises CH 2 OR6 or COOR6 of formula (1 B), wherein at least two of R1 , R2 and R6 comprise angeloyl, benzoyl, alkenoyl, or a derivative thereof.
  • R4 is a side chain comprising CH 2 OCOCH 3 , CH 2 COO-alkyl, CH 2 OH, COOH, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, or a derivative thereof.
  • R5 comprises a sugar moiety, wherein the sugar moiety comprises one or more sugar of, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, or alduronic acid: glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof.
  • R5 comprises a sugar moiety or a group capable of performing the function of the sugar moiety.
  • the R5 represents H.
  • R4 represents H, OH or CH 3 .
  • position C23, C24, C25, C26, C29 and C30 of the compound independently comprise CH 3 , CH 2 OH, CHO, COOH, COOa-lkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH 2 Oaryl, CH 2 O- heterocyclic, CH 2 O- heteroaryl, alkyls group, acetyl group or derivatives thereof, particular CH3.
  • R1 and R2 independently comprise an angeloyl group.
  • R1 is a sugar moiety or a side chain which comprise two angeloyl groups.
  • R1 and R2 independently comprise a benzoyl group.
  • R1 is a sugar moiety which is substituted with two benzoly groups.
  • R 3 represents H or OH.
  • R8 may be OH.
  • the O at C21 , 22 may be replaced by N.
  • this invention provides a method of reducing the secretion of fibronectin; wherein the medicament is for inhibiting tumor or cancer cell growth and for treating cancer, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer or cervix cancer.
  • substitution, deletion and/or addition of any group in the above- described compounds by other group(s) will be apparent to one of ordinary skill in the art based on the teachings of this application.
  • substitution, deletion and/or addition of the group(s) in the compound of the invention does not substantially affect the biological function of the compound.
  • This invention provides uses, methods, processes, compounds and compositions for modulating adhesion or angiogenesis of cancer cells, antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, inhibiting cancer metastasis or growth, reducing adhesion protein of cells, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • methods comprise inhibiting the gene expression.
  • this invention provides a method of reducing the secretion of fibronectin.
  • the method can block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer, cervix cancer, esophagus cancer, testis cancer, spleen cancer, kidney cancer, lymph cancer, pancreas cancer, stomach cancer and thyroid cancer,
  • the compounds are anti-angiogenesis, inhibiting cancer cell metastasis and inhibiting cancer growth,
  • the compounds promote angiopoietin 2 .
  • the compound is selected from the following formulas (1 E).
  • the method is administering contacting the compounds, wherein the compound is selected from the formula (1 E):
  • R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, 0-senec ⁇ oyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R4 represents CH3, CHO, CH 2 R6 or COR6, wherein R6 is selected from hydroxyl, O- angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O- alkenoyl,
  • R5 is a hydrogen, heterocyclic or sugar moiety(ies), wherein the sugar moiety(ies) is/are selected from a group consisting of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, and derivatives or combination thereof; wherein R9, R10, R1 1 , R12, R13, R14, R15 are independently attached a group selecting from CH 3 , CH 2 OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH 2 Oaryl, CH 2 O- heterocyclic, CH 2 O- heteroaryl, alkyls group,
  • R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof.
  • R5 is 3- ⁇ -O- ⁇ [( ⁇ -L-rhamnopyranosyl-(1 ⁇ 2)]- ⁇ -L-rhamnopyranosyl--(1 ⁇ 2)- ⁇ -D- galactopyranosyl--(1 ⁇ 3)]-[ ⁇ -D- galactopyranosyl--(1 ⁇ 2)]- ⁇ -D-glucuronopyranosyl ⁇ .
  • the carbon ring 3 comprises a double bond when R16 is H; wherein the double bond in carbon ring 3 is reduced when R4 and R16 form a divalent radical.
  • the method is administering contacting the compounds, wherein the compound is selected from the formula (1 F): , also named (1 F), wherein
  • R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R4 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkano
  • R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof;
  • R5 is 3- ⁇ O- ⁇ [( ⁇ -L-rhamnopyranosyl-(1 ⁇ 2)]- ⁇ -L- rhamnopyranosyl-(1 ⁇ 2)- ⁇ -D- galactopyranosyl--(1 ⁇ 3)]-[ ⁇ -D- galactopyranosyl- (1 ⁇ 2)]- ⁇ -D-glucuronopyranosyl ⁇
  • R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O
  • R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O
  • R3 is H or OH
  • R5 is H or OH
  • R6, R7, R8, R9, R10, R1 1 , R12 are independently attached a group selecting from CH 3 , CH 2 OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH 2 Oaryl, CH 2 O- heterocyclic, CH 2 O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH 3 ;
  • the use or method is administering contacting the compounds of following:
  • R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
  • R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-
  • a sugar moiety is a segment of a molecule comprising one or more sugar groups. Substitution, deletion and/or addition of any group in the above-described compounds will be apparent to one of ordinary skill in the art based on the teaching of this application. In a further embodiment, the substitution, deletion and/or addition of the group(s) in the compound of the invention does not substantially affect the biological function of the compound.
  • the method is regulating or interacting with adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
  • the method is reducing the secretion of fibronectin.
  • the method comprises inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition.
  • antiparasitics comprise inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria.
  • This invention provides a method for inhibiting the growth, migration, metastasis of cancer by altering the characteristic of membrane of cancer cell, wherein the characteristic comprise reducing adhesion protein; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK; wherein comprising inhibiting the secretion of fibronectin, wherein comprising administering to a subject, in need thereof, an appropriate amount of triterpenoidal saponins comprising two or more angeloyl groups, or a compound comprises a triterpene which comprises any two of angeloyl, tigloyl, senecioyl, perferable two angeloyl groups, and a sugar moiety, glucose, galactose, rhamnose, arabinose, xylose,
  • This invention provides a composition comprising an effective amount of the compound of any one of compound selected from the above formula or a salt, ester, metabolite or derivative thereof as a medicament for reducing adhesion protein; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK, for inhibiting the growth, migration, metastasis of cancer, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer or cervix cancer.
  • the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosy
  • This invention also provides a composition comprising the above described compounds or their derivatives for reducing adhesion protein, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK; wherein comprising inhibiting the secretion of fibronectin, wherein for treating venous insufficiency, particularly hemorrhoids or inhibiting leg swelling, or peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, Expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, episiotomies, hemonhoids, peripheral edema formation or postoperative swelling; for reducing symptoms of pain;
  • compositions comprising any one of triterpenoid saponins with the following formula: 3-0- ⁇ [/?-D-galactopyranosyl (1 ⁇ 2)]-[ ⁇ r-l_-arabinofuranosyl (1 ⁇ 3)]-/?-D- glucuronopyranoside butyl ester ⁇ -21 -0-acetyl-22-0-angeloyl-3/?,16 ⁇ ;21/?,22 ⁇ ,28- pentahydroxyolean-12-ene.
  • This invention also provides a composition for regulating or reducing adhesion protein, wherein the adhesion protein comprising fibronectin, integrins family, CD44, Myosin Vl, vitronectin collagen, laminin, Glycosylation cell surface proteins, polyglycans and FAK; inhibiting venous insufficiency, particularly hemorrhoids or inhibition of leg swelling, or inhibiting cancer growth, inhibiting leishmaniases, modulating adhesion of cancer cells, modulating angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition, comprising any of compounds selected from the following compounds (A) to (X) and (A1 ) to (X1 ) incorporated here from PCT/US2008/002086, 1 188-ALA-PCT, :
  • a triterpene comprise the following structure has activities of reducing adhesion proteins to blocks the migration, metastasis of cancer cells, growth of cancers, inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics, or manufacturing an adjuvant composition.
  • R1 , R2 and R3 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof.
  • At least one of R1 , R2 and R3 comprise a sugar moiety comprising two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof.
  • R1 , R2 or R3 comprise angeloyl groups, tigloyl groups, senecioyl groups or acetyl group or their combinations, preferable wherein at least two of the R1 , R2 and R3 comprise angeloyl groups.
  • R5 comprises sugar moiety.
  • the sugar moiety comprises at least one sugar, or glucose, or galactose, or rhamnose, or arabinose, or xylose, or alduronic acid, or glucuronic acid, or galacturonic acid, or their derivative thereof, or the combination thereof.
  • the sugar moiety comprises one or more sugar selected from, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof.
  • the sugar moiety comprise glucose, galactose or arabinose, or combination thereof, or derivatives thereof.
  • the sugar moietiy comprise alduronic acids, galactose and arabinose, wherein the alduronic comprise glucuronic acid or galacturonic acid.
  • the sugar moiety comprise alduronic acids, glucose and arabinose, wherein the alduronic comprise glucuronic acid or galacturonic acid.
  • R5 is Hydrogen.
  • the R1 , R 2 and R3 may be attached in other position of the structure.
  • the compound is thterpenoid saponin comprise comprises at least two angeloyl groups, tigloyl groups, senecioyl groups or acetyl group or their combinations, preferable wherein at least two angeloyl groups.
  • At least one of the side bonds comprise a sugar moiety comprising two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof.
  • the compound comprises a sugar moiety.
  • the sugar moiety comprises glucose, galactose or arabinose or combination thereof.
  • the sugar moiety comprises at least one sugar, or glucose, or galactose, or rhamnose, or arabinose, or xylose, or alduronic acid, or glucuronic acid, or galacturonic acid, or their derivative thereof, or the combination thereof.
  • the sugar moiety comprises one or more sugar selected from, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof.
  • sugar selected from, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof.
  • a composition comprising an effective amount of compound selected from the above formula or a salt, ester, metabolite or derivative thereof as a medicament for regulating or reducing adhesion protein, blocking the migration, metastasis of cancer cells, inhibiting tumor or cancer cell growth and for treating cancer, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal and cervix cancer.
  • a compound or sapongenin comprises the structure (d) or (e) has anti-cancer or inhibiting virus activities.
  • a composition for regulating or reducing adhesion protein, blocking the migration, metastasis of cancer cells, treating cancers or inhibiting virus comprising a compound, wherein the compound is a triterpene, which comprises at least two side chains which comprise angeloyl groups, wherein the side chains are at adjacent carbon in trans position.
  • the side chains are at alternate carbon in cis position.
  • the side chains are at alternate carbon in trans position.
  • the side chains are attached an acyl.
  • the side chains are attached an unsaturated group.
  • the side chains are in non-adjacent carbon cis or trans position.
  • the side chains comprise a functional group capable of performing the function of angeloyl group.
  • the above compounds can be used for regulating or reducing adhesion protein, blocking the migration, metastasis of cancer cells, inhibiting tumor cell growth, reducing leg swelling, symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, by administering to a subject in need thereof, an effective amount of the above described compounds.
  • This invention provides a method for inhibiting tumor cell growth, regulating cell growth, reducing inflammation, inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition, comprising administering to a subject, in need thereof, an effective amount of the compound which comprises any of the above structures to said subject.
  • the cancers are included but not limited to Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma cancer, Ovarian cancer, Renal cancer, Prostate cancer, Breast cancer, bladder cancer, cervix cancer, liver cancer, bone cancer, brain cancer and Skin cancer.
  • This invention also provides a method for reducing swelling, reducing symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, Expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, for reducing symptoms of pain; thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, comprising administering to a subject, in need thereof, an effective amount of the composition of this invention.
  • This invention provides a composition comprising the compounds provided in the invention for treating cancers; for inhibiting virus; for preventing cerebral aging; for improving memory; improving cerebral functions, for curing enuresis, frequent micturition, urinary incontinence, dementia, Alzheimer's disease, autism, brain trauma, Parkinson's disease or other diseases caused by cerebral dysfunctions; for treating arthritis, rheumatism, poor circulation, arteriosclerosis, Raynaud's syndrome, angina pectoris, cardiac disorder, coronary heart disease, headache, dizziness, kidney disorder; cerebrovascular diseasea; inhibiting NF-Kappa B activation; for treating brain edema, sever acute respiratory syndrome, respiratory viral diseases, chronic venous insufficiency, hypertension, chronic venous disease, anti-oedematous, anti inflammatory, hemonhoids, peripheral edema formation, varicose vein disease, flu, post traumatic edema and postoperative swelling;for inhibiting blood clot, for inhibiting ethanol absorption; for lowering blood
  • This invention provides a composition for AntiMS, antianeurysm, antiasthmatic, antibradykinic, anticapillarihemorrhagic, anticephalagic, anticervicobrachialgic, antieclamptic, antiedemic, antiencaphalitic, antiepiglottitic, antiexudative, antiflu, antifracture, antigingivitic, antihematomic, antiherpetic, antihistaminic, antihydrathritic, antimeningitic, antioxidant, antiperiodontic, antiphlebitic, antipruritic, antiraucedo, antirhinitic, antitonsilitic, antiulcer, antivaricose, antivertiginous, cancerostatic, corticosterogenic, diuretic, fungicide, hemolytic, hyaluronidase inhibitor, lymphagogue, natriuretic, pesticide, pituitary stimulant, thymolytic, vasoprotective, inhibiting leishmaniases, modulating
  • Alkenyl means unsaturated linear or branched structures and combinations thereof, having 1 -7 carbon atoms, one or more double bonds therein.
  • alkenyl groups include vinyl, propenyl, isopropenyl, butenyl, s- and t-butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, and hexadienyl.
  • An aryl is a functional group of organic molecule derived from an aromatic compound such as benzene, a 6-14 membered carbocyclic aromatic ring system cpmprising 1 -3 benzene rings. If two or more aromatic rings are present, then the rings are fused together, so that adjacent rings share a common bond. Examples include phenyl and naphthyl.
  • the aryl group may be substituted with one or more sunstitutes independnetly selected from halogen, alkyl or alkoxy.
  • Acyl is a functional group obtained from an organic acid by the removal of the carboxyl.
  • Acyl groups can be written as having the general formula -COR, where there is a double bond between the carbon and oxygen.
  • the names of acyl groups typically end in -yl, such as formyl, acetyl, propionyl, butyryl and benzoyl.
  • Benzoyl is one of acyls, C 6 H 5 COR, obtained from benzoic acid by the removal of the carboxyl.
  • Heterocyclic compound - a compound containing a heterocyclic ring which refers to a non-aromatic ring having 1 -4 heteroatoms said ring being isolated or fused to a second ring selected from 3- to 7-membered alicyclic ring containing 0-4 heteroatoms, aryl and heteroaryl , wherein said heterocyclic comprises pyrrolidinyl , pipyrazinyl , morpholinyl , trahydrofuranyl, imidazolinyl, thiomorpholinyl , and the like.
  • Alkanoyl is the general name for an organic functional group RCO-, where R represents hydrogen or an alkyl group.
  • R represents hydrogen or an alkyl group.
  • alkanoyl is selected from acetyl, propionoyl, butyryl, isobutyryl, pentanoyl and hexanoyl.
  • Alkenoyl is alkenylcarbonyl in which alkenyl is defined above. Examples are pentenoyl(tigloyl) and hexenoyl(angeloyl).
  • Alkyl is a radical containing only carbon and hydrogen atoms arranged in a chain, branched, cyclic or bicyclic structure or their combinations, having 1 -18 carbon atoms. Examples include but are not limited to methyl, ethyl, propyl isopropyl, butyl, s- and t- butyl, pentyl, hexyl, heptyl, octyl, nonyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • Benzoyl alkyl substituted alkanoyl is refer to straight or branched C1 -C6 alkanoyl substituted with at least one benzoyl and at least one alkyl, wherein the benzoyl is attached to a straight or branched C1 -C6 alkyl.
  • a benzoyl alkyl substituted alkanoyl is benzoyl methyl isobutanoyl.
  • a sugar moiety is a segment of molecule comprising one or more sugars or derivatives thereof or alduronic acid thereof.
  • Benzoyl is [0-C2 alkanoyl substituted with phenyl]
  • This invention provides a composition and method for inhibiting the growth, migration, metastasis of cancer by altering the adhesion characteristic of membrane of cancer cell, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer or cervix cancer, wherein the method is administering contacting Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, or a salt, ester, metabolite thereof. In an embodiment the method is administering contacting the compound selected from formula in this application.
  • Xanifolia-Y is an alternate or supplemental agent to DNA- inhibition or microtubule-targeting drugs. It could be beneficial if it is used singly or in combination with other drugs of different mechanisms (block M-phase progression or DNA synthesis).
  • Our inventions show combined effect of Xanifolia-Y and paclitaxel on inhibition of ES2 cells' growth (Detail in Experiment 14 U.S. Serial Nos.1 1/683198, filed on March 7, 2007) .
  • Xanifolia-Y extended the life span of tumor bearing mice. (Reference Experiments 7, 8, 9 in U.S. Serial Nos.1 1/683198, filed on March 7, 2007,). The animals died sooner if the treatment of Xanifolia-Y was delayed (comparing results of treatments started from 1 , 4 or 10 days after tumor inoculation). The results show that Xanifolia-Y inhibits migration or metastasis of the inoculated cancer cells. Ovarian carcinoma cells express high levels of adhesion molecules. Adhesion proteins are present in both cancer cells and mesothelial cells.
  • Fibronectin is a kind of glycoprotein that binds to membrane spanning receptor proteins comprising the integrins, collagen, fibrin and heparin sulfate. Fibronectin has been implicated in tumor development and metastasis.
  • This application provides methods and compositions for modulating the gene expression of fibronectin, inhibiting the secretion of fibronectin, reducing the receptors of fibronectin, reducing the adhesion ability fibronectin, inhibiting the metastasis, or inhibiting cancer growth, wherein the method and composition comprises administering to the said subject as effective amount of compounds selected in this appliaction.
  • Angiogenesis is a process involving the growth of new blood vessels. It is a normal process in growth and development. However, this is also a fundamental step in the transition of tumors from a dormant state to a malignant state.
  • the angiopoietins are protein growth factors that modulate angiogenesis.
  • the identified angiopoietins comprise angiopoietin 1 , angiopoietin 2, angiopoietin 3, angiopoietin 4, angiopoietin 5, angiopoietin 6, angiopoietin 7, angiopoietin-like 1 , angiopoietin-like 2, angiopoietin-like 3, angiopoietin-like 4, angiopoietin-like 5, angiopoietin-like 6, and angiopoietin-like 7.
  • the angiopoietin 1 is a positive foctor to promote the new blood vessels.
  • the angiopoietin 2 is antagonist of angiopoietin 1 , which is a negative factor for the growth of new blood vessels.
  • This application provides methods and compositions for modulating angiopoietin and inhibiting cancer growth; wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal and cervix cancer, wherein the methods and compositions comprise administering to the said subject as effective amount of compounds selected in this appliaction.
  • the compounds in this application are positive regulating angiopoietin 2.
  • the compounds in this application are negative regulating the angiopoietin 1 .
  • the results of the micro array experiment showed that compound Y and YM (ACH-Y) modulate the gene expression of angiopoietin family in ES2 cells. They promote angiopoietin 2 and inhibit angiopoietin 1 and angiopoietin-like 1 and angiopoietin-like 4.
  • the compounds in this application are used antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, anti-angiogenesis, inhibiting cancer cell metastasis and inhibiting cancer growth, wherein the compounds comprise Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, ACH-Y or a salt, ester, metabolite thereof and compounds selected from formula (1 A), (1 B), (1 C) , (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof.
  • the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D) (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L).
  • the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety.
  • the compound(s) are selected from Compound A to X and A1 to X1 in the application.
  • the compound(s) are selected from Compound Z1 to Z7 in the application.
  • the compound(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E.
  • the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba 17.
  • the triterpene compounds of this invention can be administered to a subject in need thereof treating the subject, wherein including preventing canceror has adjuvant effect to the subject, or inhibiting the initation or promotion of cancer, or killing the cancer/tumor cells.
  • the compounds inhibit the activation of nuclear factor-kB, wherein inhibiting the localization or wherein binding the DNA.
  • the compounds induce apoptosis in cancer cells.
  • the triterpene compounds of this invention can reduce blood vessel in the tumor in a subject. ( Figure 1 )
  • the results of the micro array experiment showed that compound Y and YM inhibit gene expression of insulin-like growth factor binding protein in ES2 cells.
  • F1 1 For lung carcinoma cells (H460), at concentration of 20ug/ml, there are inhibitions of Fibronectin secretion ranged from 20-60%.
  • F12A For bladder carcinoma cells (HTB-9), Xanifolia-Y (10 ug/ml) inhibits 50% of
  • Angiopoietin 2 (Ang2) Up regulation of Angiopoietin 2 (Ang2) in ES2 cells with Xanifolia-Y treatment.
  • APEH APEH, APLP2, ARL10C, ARMC4, ARMCX1 , ARMCX6, ARNTL2, ARNTL2, ATF3,
  • ATP6V0E ATP6V1 B2, ATP6V1 C1 , ATP6V1 C1 , BCL2A1 , BCL6, BRI3, BTD, C14orf109, C14orf78, C17orf32, C6orf65,C9orf10, C9orf103, CAD, CAV1 , CAV2, CBLL1, CCL20, CD33L3, CEBPB, CEP4, CFH /// CFHL1, CHRDL1, CITED2, CITED2, CLDN14, CLN8, CLTA, CNAP1, COG6, COL18A1, COL4A2, COL5A1, COL5A2, COL6A3, COPG, CPM, CPNE3, CPSF1, CSRP2BP, CSTB, CTNS, CXCL2, DDB1, DDIT3, DDX20, DKFZP564I1171 , DKFZP586J0619, DUSP10, DUSP10, DY
  • P8 KRTAP4-7, DUSP10, CLDN14, ANGPT2, DUSP10, GDF15, GPNMB, HDAC9, HECW2, C14orf78, UPP1, PPP1R15A, PLEKHM1, STX3A, ANGPT2, SQSTM1, RHOQ, STC2, PPP1R15A, LOC286044, ATF3, HMOX1, CXCL2, CD33L3, LRRC8E, SESN2, LIF, TFPI2, KIAA1706, RRAGC, DDIT3, DYRK3, CTNS, GPNMB, CEBPB, CCL20, AMIGO2, KIAA1462, HSPBAP1, EVC, CLN8, ABL2, SGEF, MCTP1, IRS2, C14orf109, TNFAIP3, RND3, ZFHX1 B, LHFPL2, SNF1LK, SLC3A2, NAV2, SPOCD1, TFPI2, EVI2A, ST3GAL6, CSTB
  • ANGPT2 angeopoietin 2
  • LIF Leukemia inhibitory factor
  • ITGAV integrin alpha V, (cholinergic differentiation factor),
  • COL1 A1 collagen type 1 alpha 1 , polypeptide gene enhancer in B-cells
  • FN1 fibronectin 1 , inhibitor, zeta
  • LAMA4 laminin alpha 4
  • DDIT3 DNA-damage-inducible transcript
  • Xanifolia Y modulates gene expression.
  • This invention provides compositions and methods for modulating the gene expression in cancer cells, wherein the modulating comprises of positive and negative regulation, wherein genes being modulatated are adhesion proteins; wherein modulation includes expression, production and secretion of adhesion proteins, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, cadherin, heparin, Glypican, tenascin, CD 54, CAM.
  • compositions and methods for modulating angiopoietins wherein comprises positive regulating the angiopoietin 2, wherein comprises negative regulating angiopoietin 1 .
  • the composition and method of this invention comprises a triterpene wherein acylation group at carbon position 21 and/or 22 of the triterpene is necessary for the function and are selected from angeloyl, acetyl, alkanoyl, and alkenoyl and acyl group.
  • the sugar moiety (ies) at position 5 of the triterpene is important for enchancing activity of these compounds.
  • Leptin is a promoter in breast cancer cells.
  • the Xanifolia Y and YM inhibits the expression of leptin.
  • the thrombospondins are a family of multifunctional proteins.
  • the family includes thrombospondins 1 -5.
  • Thrombospondin-1 TSP1 is a multifunctional, matricellular glycoprotein, containing interacting domains for a large variety of adhesion, migration and proliferation, angiogenesis and tumor cell metastasis.
  • the insulin-like growth factor binding protein is a multifunctional protein that relate to metastasis, tumor growth and angiogenesis.
  • Compounds in this application modulate gene expression of RAB3B, inhibiting metastasis, tumor growth and angiogenesis.
  • RAB3B is member RAS oncogene family protein. Compounds in this application modulate gene expression of RAB3B, inhibiting metastasis, tumor growth and angiogenesis.
  • the saponin can be partially hydrolyzed into a mixture of products which are separated by HPLC. Specific partial hydrolysis of saponins can also be achieved with enzymes.
  • the glycosidases catalyze the hydrolysis of the glycosidic linkage.
  • Galactosidase is an enzyme which catalyzes the hydrolysis of galactosides.
  • the glucosidase is an enzyme which breaks glucose from saponin.
  • Other enzyme examples are xylanases, lactase, amylase, chitinase, sucrase, maltase, and neuraminidase.
  • the sugar moiety of the triterpenoid saponin can be removed by acid hydrolysis.
  • the synthetic compound of ACH-Y is obtained.
  • the ACH-Y is a triterpene with acyl groups but no sugar moiety.
  • the acyl group of the saponin (example Xanifolia Y) can be removed by alkaline hydrolysis.
  • a synthetic compound of AKOH-Y can be obtained.
  • the AKOH-Y is a pentacyclic triterpene with sugar moieties.
  • a pentacyclic triterpene can be obtained by acid and alkaline hydroysis of saponin from natural source.
  • a pentacyclic triterpene also can be obtained by other methods (Reference: Surendra et al., Rapid and Enantioselective Synthetic Approches to Germanicol and Other Pentacyclic Triterpenes).
  • the pentacyclic triterpene with sugar moieties can also be obtained with synthesis (Reference: PIe et al., Synthesis of L- arabinopyranose containing hederagenin saponins).
  • Acylation is the process of adding an acyl group to a compound. Friedel-Crafts reaction is an example of the process.
  • An active compound can be obtained by acylating a pentacyclic triterpene.
  • acylating G21 and C22 of a pentacyclic triterpene gives an anticancer compound .
  • sugar moiety(s) at C3 can increase the activities of pentacyclic triterpene, wherein the triterpene has acyl group(s), wherein the acyl group(s) may be at C21 , 22, or 28.
  • a sugar moiety is at C21 , 22, or 28, wherein the sugar moiety is substituted with 2 acyl groups.
  • Y10 is cytotoxic to Leishmania Major (promastigotes) with IC50 of 15ug/ml.
  • IC 50 of Y is 15ug/ml
  • YO is 25ug/ml
  • Y1 is 23ug/ml
  • Y5 is 16ug/ml
  • Y7 is 18ug/ml
  • Mb5 is 15ug/ml
  • ACH-Mb5 is 18ug/ml
  • Z12 is 23ug/ml
  • Ba1 is 15ug/ml
  • the solution was incubated in 8OC water bath for 4 hours. It was cooled to room temperature before neutralized with 0.5 ml 1 N HCI (adjust pH to about 3). The mixture was extracted with 2 ml 1 -butanol 3 times. The butanol fractions were collected and lyophilized. The hydrolyzed saponin with further purified with HPLC in a C-18 column eluted with 25% acetonitrile.
  • Athymic Nu/Nu mice (5-6 weeks old) are divided into two groups (1 and 2) with 5 animals in each group. On day 0, all mice were transplanted intra-peritoneally with one million ES2 (human ovarian cancer) cells. Animals were randomly divided into two groups: Group 1 : Control group. Animals did not receive drug-treatment.
  • Group 2 Drug-treatment group. On days 10-15 and 18-22, animals received daily drug administration of Xanifolia-Y, by i.p. route at dosage of 2.5 mg/kg.
  • mice showed high tumor burden after 18 days. Animals with high tumor burden were euthanized and solid tumors were taken out from these mice (between 18-
  • Leishmania parasites (Leishmania major: MRHO/SU/59/P/LV39) were grown in culture medium in a T75 flask at room temperature. Promastigotes of Leishmania major (approximately 40 million per ml) were used in the experiment. 1 .2 ml cell culture was transferred to a well of the 24-wells plate. Saponin Y10 (0.2 ml in medium) with different concentrations (final 6.25 -200 ug/ml) was added to culture and cells were grown for 1 -5 days at room temperature. At the end of drug-treatment, 150 ul of MTT (5 mg/ml in PBS) was added to each well and incubated for 4 hours.
  • MTT 5 mg/ml in PBS

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Cosmetics (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)

Abstract

Methods for utilizing the anticancer properties of terpenoid saponin compounds so as to modulate adhesion proteins, inhibit angiogenesis in tumors, modulate gene expression, modulate angiopoietin, enhance an immune response, provide adjuvant activities, provide vaccine activities, or inhibit the metastasis of cancer cells when contacting a subject with the compounds, wherein the cancers compose breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer, cervix cancer, esophagus cancer, testis cancer, spleen cancer, kidney cancer, lymph cancer, pancreas cancer, stomach cancer and thyroid cancer Methods, processes, compounds and compositions for antiparasitic applications which include inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria

Description

INTERNATIONAL PATENT APPLICATION UNDER THE PATENT COOPERATION TREATY
To all whom it may concern:
Be it known that Pui-Kwong CHAN and May Sung MAK have invented certain new and useful improvements in
BLOCKING THE METASTASIS OF CANCER CELLS AND THE USES OF NEW COMPOUNDS THEREOF of which the following is a full, clear and exact description, BLOCKING THE METASTASIS OF CANCER CELLS AND THE USES OF NEW COMPOUNDS THEREOF
This application claims benefit of U.S. Serial No. 61/038,277 filed March 20, 2008, U.S. Serial No. 61/054,308, filed May 19, 2008, International App'l No. PCT/US2008/002086, filed February 15, 2008, International App'l No. PCT/US2007/077273, filed August 30, 2007, U.S. Serial No. 60/890,380, filed on February 16, 2007, U.S. No. 60/947,705, filed on July 3, 2007, and U.S. Serial No.1 1/683,198, filed on March 7, 2007, which claims benefit of U.S. Serial Nos. 60/795,417, filed on April 27, 2006, 60/841 ,727, filed on September 1 , 2006, 60/890,380, filed on February 16, 2007, and International Application No. PCT/US2006/016158, filed April 27, 2006, which claims the benefit of the priority of the following applications: (1 ) U.S. Serial Nos. 1 1/289142, filed November 28, 2005, and 1 1/267,523, filed November 4, 2005; (2) International Application No. PCT/US05/31900, filed September 7, 2005 (which claims the priority of U.S. Serial Nos. 60/617,379, filed October 8, 2004, 60/613,81 1 , filed September 27, 2004, and 60/607,858, filed September 7, 2004); (3) U.S. Serial No. 1 1 /131 ,551 , filed May 17, 2005; and (4) U.S. Serial No. 1 1/1 17,760, filed April 27, 2005. This application also claims benefit of U.S. Serial No.1 1 /412,659, filed April 27, 2006, U.S. Serial No. 10/906,303, filed February 14, 2005, and U.S. Serial No. 12/344,682, filed December 29, 2008. The contents of these preceding applications are hereby incorporated in their entireties by reference into this application.
FIELD OF THE INVENTION
This invention provides methods and compositions for reducing the adhesion protein in cell and blocks the migration, metastasis of cancer cells or inhibits the growth of cancers or anti-angiogenesis; wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers, wherein the adhesion proteins or receptors comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
BACKGROUND OF THE INVENTION
The composition from Maesa balansae for anti-leishmanial is a United Nation funding project. This invention is providing methods for inhibiting cancer metastasis or growth, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers, wherein the method is using the extract, composition and compounds from Maesa balansae, Barringtonia acutangula, Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum.
SUMMARY OF THE INVENTION
This invention is providing methods of using the extract, composition and compounds from Maesa balansae and Barringtonia acutangula, Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum for reducing adhesion protein, for inhibiting cancer metastasis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers.
This invention provides a method for modulating the adhesion of cancer cell and blocking their migration, metastasis or inhibiting the growth of cancers or anti- angiogenesis, wherein the adhesion protein and their receptors comprise fibronectin, integhns family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
This invention provides a method of antiprotozoal comprising treating leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis and malaria.
DETAILED DESCRIPTION OF THE FIGURES
Figure 1 : Analysis of genesis of blood vessel in xenograft tumor treated with compound Y. Figure A and B show the tumor sections taking from mice without Xanifolia Y treatment. Figure C and B show the tumor sections taking from mice with Xanifolia Y treatment. More blood vessels were observed in the control Groupi than those in the drug-treated Group 2
Figure 2: Experiment shows that Y10 is cytotoxic to Leishmania Major (promastigotes) with IC50 approximately equal to 15ug/ml.
Figure 3: Shows the inhibition activity of compound Y, X, ACH-Y and AKOH-Y
DETAILED DESCRIPTION OF THE INVENTION
This invention provides methods and compositions for modulating the gene expression to cure diseases or reduce the syndrome of diseases, wherein the modulating comprises positive and negative regulating. In an embodiment, the method comprises inhibiting the gene expression. In an embodiment the method comprises stimulating the gene expression.
This invention provides methods and compositions for inhibiting the migration, metastasis or growth of cancers or anti-angiogenesis, wherein the methods comprise affecting the gene expression, wherein comprise affecting the adhesion proteins or their receptors, reducing adhesion protien, or inhibiting the expression or secretion of adhesion proteins, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
This invention provides methods and compositions for inhibiting the migration, metastasis or growth of cancers or anti-angiogenesis, wherein the methods comprise affecting the gene expression, wherein comprises stimulating the gene expression.
This invention provides a method for altering the characteristic of cancer cell membrane resulting in blocking the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the method comprises reducing adhesion protiens or their receptors, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
This invention provides methods, processes, compounds and compositions of reducing adhesion protein of cells, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD54, CAM, elastin and FAK. In an embodiment, methods comprise inhibiting the gene expression. In an embodiment, this invention provides a method of reducing the secretion of fibronectin. In an embodiment the method can block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer. In an embodiment, the method is contracting the said cell with compound selected from Mb1 , Mb2, Mb2.1 , Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13, ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba17, Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof and Compound A to X and A1 to X1 in the application.
This invention provides a method of altering the characteristic of cancer cell membrane, wherein the method comprises altering the secretion of adhesion proteins, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, the methods, processes, compounds and compositions comprises blocking, suppressing or inhibiting the expression or secretion of adhesion protein, wherein the adhesion proteins . In an embodiment, the methods, processes, compounds and compositions is interacting with adhesion protein, wherein the adhesion proteins. In an embodiment the methods, processes, compounds or compositions can block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer. In an embodiment, the method is contracting the said cell with compound selected from Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13,, ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba 17, Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof and Compound A to X and A1 to X1 in the application.
The adhesion proteins help cancer cell adhesion, invasion or metastasis, wherein the cancers comprise ovarian cancer. Reducing the adhesion proteins will reduces the metastasis of cancers.The fibronectin is one of the key factors in the biology of epithelial ovarian cancers. The reducing of fibronectin will inhibit the metastasis of cancer cells.
This invention provides a method and composition for inhibiting the secretion of adhesion protein comprising fibronectin in order to cure the diseases, wherein the diseases comprise inhibiting cancer growth, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer. In an embodiment, the method is contracting the said cell with compound selected from Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, MbI O1 Mb1 1 , Mb12 and Mb13, ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5, ACH-MbI 3, Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba 17, Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof and Compound A to X and A1 to X1 in the application.
This invention provides a use of compound for manufacture of medicament or a composition for inhibiting the growth, migration, metastasis of cancer and by altering the characteristics of membrane of cancer cell, wherein the characteristics comprise adhesion of proteins; wherein comprising the secretion of proteins or the adhesion of cells; wherein the characteristic comprise adhesion ability; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK; wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer; wherein the method is administering contacting the say cells with the extract, compositions, saponins and compounds from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and Barringtonia acutangula for inhibiting cancer metastasis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers; wherein extracts, compositions, saponins and compounds are prepared from the husks, branches, stems, leaves, kernels, roots, barks, fruit, seeds or seed shells of the herb or plant.
The present invention provides vaccine for cancer immunotherapy. The vaccines comprise extract, compositions, compounds and saponins from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula. In embodiment, the compounds can be obtained from synthesis, semi-synthesis or modification. The method comprises administering to the said subject and effective amount of vaccine for enhancing the immune response. The vaccines comprise saponins isolated from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula.
The present invention provides adjuvant compositions for cancer curing. The adjuvant compositions comprise extract, compositions, compounds and saponins from
Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or
Barringtonia acutangula. The method comprises administering to the said subject and effective amount of the above adjuvant compositions for enhancing the immune response. The use of vaccine compositions comprise inhibiting cancer metastasis, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers.
The present invention provide a use of compound for manufacture of medicament or methods for making vaccine, wherein the vaccine comprise compounds or saponins from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula, wherein the vaccine can be used for inhibiting cancer growth, wherein the vaccine is having immune adjuvant activity, wherein the saponins comprise Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment the saponin may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the saponin comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13,. In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the compound(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5, and ACH-MbI 2. In an embodiment the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16 and Ba 17. In an embodiment the compound(s) are selected from Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment, this invention provides a method for antiparasitics by using the above compounds, wherein the antiparasitics comprise inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria; wherein a method comprises contacting cells with an effective amount of an isolated, purified or synthesized compound, or its salt, or ester thereof, selected from the above compounds.
This invention provides a use of compound for manufacture of medicament or a method for pharmaceutical composition useful for inducing an immune response to and antigen in an individual comprising the saponin composition from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae or Barringtonia acutangula or synthesis. The present invention provide methods for enhancing an immune response to an antigen in an individual comprising administering an effective amount of saponins/compositions from Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae and/or Barringtonia acutangula, or the saponin comprise Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof, or the saponin may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the saponin comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13, In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the composition(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5, and ACH-MbI 2. In an embodiment the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba17.
This invention is providing methods or a use of compound for manufacture of medicament for modulating adhesion or angiogenesis of cancer cells, antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, inhibiting cancer metastasis or growth, of using the extract, composition and compounds from Maesa balansae, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancers; In an embodiment the method is modulating the phosphatidylinositol signaling system, regulate the gene expression of RGS4 , LEPR, ICFBP3, ANGPT2, GPNMB, NUPR1 or LOC100126784. The compounds can be purified from natural resource comprising Xanthoceras Sorbifolia, Harpullia, Aesculus hippocastanum, Maesa balansae or Barringtonia acutangula, or synthesized. The compounds comprise following: (Our purification methods and biological assays including the MTT assay in International Application No. PCT/US05/31900, filed September 7, 2005, U.S. Serial No. 1 1 /289142, filed November 28, 2005, and U.S. Serial No. 1 1/131551 , filed May 17, 2005, and PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008, the contents of which are incorporated herein by reference)
wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is (E) O(C=O)CH=CH-C6H5, R3 is OH, R4 is OH, also named Mb1 ; or wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is (Z)- O(C=O)CH=CH-C6H5, R3 is OH, R4 is OH, also named Mb2; or wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is (Z)-O(C=O)C(CH3)=CH-C6H5, R3 is OH, R4 is OH, also named Mb2.1 ; or wherein R1 is O(C=O)C6H5, R2 is (E)-O(C=O)CH=CH-C6H5, R3 is OH, R4 is OH, also named Mb3; or wherein R1 is O(C=O)C6H5, R2 is (Z)- O(C=O)CH=CH-C6H5, R3 is OH, R4 is OH, also named Mb4; or wherein R1 , R2, are O(C=O)C(CH3)=CH(CH3), R3 is OH, R4 is OH, also named Mb5; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)C(CH3)=CH(CH3), R3 and R4 are OH, also named Mb6; or
wherein R1 is O(C=O)C6H5, R2 is O(C=O)CH=CH-C6H5, R3, R4, R5 are OH, R6, R7,R8, R9, R10, R1 1 , R12 are CH3, R13 is COOH also named Mb7; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)CH=CH-C6H5, R3, R4, R5 are OH, R6 is CH2OH, R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOH also named Mb8; or wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is O(C=O)CH=CH-C6H5, R3, R4 are OH, R6 , R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOH also named Mb9; or wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is O(C=O)CH=CH-C6H5, R3, R4 are OH, R6 is CH2OH, R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOH, also named Mb10; or wherein R1 , R2, are O(C=O)C(CH3)=CH(CH3), R3 is OH, R4 is CH2OH, R5 is H, R6, R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOH, also named Mb1 1 ; or wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is O(C=O)CH=CH-C6H5, R3, R4 are OH, R6, R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOCH3, also named Mb12; or wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is (Z)-O(C=O)C(CH3)=CH-C6H5, R3 is OH, R4 is OH, R5 is H, and R6, R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOH, also named Mb13; or
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-angeloyl, 22-O-angeloyl-3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba 1
wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is O(C=O)C(CH3)=CH(CH3), R3 is
COOCH3, formula is 3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- methylglucuronopyranosyl-21 -O-angeloyl, 22-O-angeloyl-3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba 2; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)C(CH3)=CH(CH3), R3 is COOH, formula is: 3-O-[β-D-galactopyranosyl(1 →2)]- β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-21 -O-benzoyl, 22-O-angeloyl-3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba 3; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)C(CH3)=CH(CH3), R3 is COOCH3, formula is: 3-O-[β-D- galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-methylglucuronopyranosyl-21 -O- benzoyl, 22-O-angeloyl-3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba 4; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)C6H5, R3 is COOH, formula is: 3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-benzoyl, 22-O-benzoyl-3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba 5; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)C6H5, R3 is COOCH3, formula is: 3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- methylglucuronopyranosyl-21 -O-benzoyl, 22-0-benzoyl-3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba 6; or wherein R1 is O(C=O)C6H5, R2 is O(C=O)CH3CH3, R3 is COOH, formula is:
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-benzoyl, 22-O-isobutyryl-3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba 7; or wherein R1 is O(C=O)C6H5, R2 is OH, R3 is COOH, formula is:
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-benzoyl -3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba 8
wherein R1 is OH , R2 is O-benzoyl, R3 is O-benzoyl,
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-[3,4-dibenzoyl-β-D-xylopyranosyl]- 3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12- ene, named Ba9
R1 is O-acetyl, R2 is O-benzoyl, R3 is O-benzoyl 3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-[3,4-dibenzoyl-β-D-xylopyranosyl]-22-O-acetyl- 3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba10
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-[3-angeloyl, 4-benzoyl--β-D-xylopyranosyl]- 3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba1 1
3-O-[β-D-galactopyranosyl(1 ^2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-[3,4-diangeloyl--β-D-xylopyranosyl]- 3β, 15α, 21 β, 22α, 28-pentahydroxyolean-
12-ene, named Ba12 3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-[3-angeloyl, 4-tigloyl-β-D-xylopyranosyl]- 3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba13
Wherein R1 is OH , R2 is O-benzoyl, R3 is O-benzoyl
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-[3,4-dibenzoyl--α-L-arabinopyranosyl]- 3β, 15α, 21 β, 22α, 28-pentahydroxyolean-
12-ene, named Ba14
Wherein R1 is O-acetyl, R2 is O-benzoyl, R3 is O-benzoyl
3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-
21 -O-[3,4-dibenzoyl--α-L-arabinopyranosyl]-22-O-acetyl- 3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba15
,3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-21 -O-[3-angeloyl-4-(3-benzoyl-2-methylbutyryl)-α-L- arabinopyranosyl]-22-O-acetyl- 3β, 15α, 21 β, 22α, 28-pentahydroxyolean-12-ene, named Ba16
;3-O-[β-D-galactopyranosyl(1 →2)]-β- D- xylopyranosyl (1 →3)-β-D-glucuronopyranosyl-21 -O-angeloyl, 22-O-angeloyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene, also named Ba 17 Anti-cancer activities:
The anti-cancer activity of Mb's compounds with ES2 cell: the IC50 of Mb1 is 8ug/ml, Mb2 is /ml, Mb3 is 8ug/ml, Mb4 is 15ug/ml, Mb5 is 6.5ug/ml, and Mb6 is 10ug/ml, Mb7 is 12ug/ml, Mb8 is 20ug/ml, Mb9 is 18ug/ml, Mb12 is 10ug/ml.
The anti-cancer activity of ACH's compounds with ES2 cell: the IC50 of ACH-Z4 is 40 ug/ml, ACH-Y3 is 20 ug/ml, ACH-Y10 is 20 ug/ml, ACH-Y2 is 35 ug/ml, ACH-Y8 is 35 ug/ml, ACH-Y7 is 65 ug/ml, ACH-YO is 20 ug/ml, ACH-X is 40ug/ml, ACH-E is 60 ug/ml The anti-cancer activity of Ba compounds with ES2 cell: Ba1 is 5 ug/ml, Ba2 is 5 ug/ml, Ba3 is 8 ug/ml, Ba 5 is 16 ug/ml, Ba7 is 1 1 ug/ml, Ba8 is 20 ug/ml, Ba9 is 12 ug/ml, Ba17 is 5 ug/ml.
The anti-cancer activity of Xanifolia Y with cells: the IC50 of Y on (bladder)TB9 cells is 5 ug/ml; IC50 of Y on (lung)H460 cells is 7.5 ug/ml; IC50 of Y on HeLa cells is 20 ug/ml; IC50 of Y on skin cells is 12 ug/ml ; IC50 of Y on ES2(ovarian) cells is 5 ug/ml; IC50 of pure Y on (Mouth) KB cells is 6 ug/ml; IC50 of Z12 on ES2(ovarian) cells is 16 ug/ml; Z4 is 20ug/ml
IC50 of Mb5: (bladder)TB9 is 6.5ug/ml, (Prostate) DU145 is 7.6 ug/ml, (Lung) H460 is 12 ug/ml (Liver) HepG2 is 6.5 ug/ml, (brain) T98G is 12 ug/ml, (Skin) SK-MELS is 25 ug/ml, (Ovary)ES2 is 6.5 ug/ml, (Breast)MCF7 is 1 1 ug/ml.
IC 50 of ACH-Mb5: (bladder)TB9 is 5.7ug/ml, (Prostate) DU145 is 6.4 ug/ml, (Lung) H460 is 6.5 ug/ml (Liver) HepG2 is 4 ug/ml, (brain) T98G is 6 ug/ml, (Skin) SK-MELS is 22 ug/ml, (Ovary)ES2 is 8 ug/ml, (Breast) M C F7 is 13 ug/ml.
This invention provides a method and composition for reducing of adhesion protein to cure the diseases, wherein the diseases comprise inhibiting cancer growth, reducing leg swelling, symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, for reducing symptoms of pain; thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, comprising administering to a subject, in need thereof, an effective amount of the composition of this invention. In an embodiment, the method comprises interacting with adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, this invention provides a method of reducing the secretion of fibronectin.
In an embodiment, the method comprises reducing the adhesion ability of adhesion protein; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK.
In an embodiment, the method comprises modulating the secretion of adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, the method comprises blocking the secretion of adhesion protien, wherein the adhesion protein comprising fibronectin. In an embodiment the method is administering contacting an effective amount of the compound selected from formulas in this application.
In an embodiment, this invention provides a method and composition for modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition. In an embodiment, this invention provides a method of antiprotozoal comprising treating leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis and malaria.
This invention provides a use of compound for manufacture of medicament, a method and composition for altering the characteristic of adhesion protein to cure diseases, wherein the characteristic comprising adhesion ability, wherein the method comprises reducing the secretion of fibronectin, wherein the diseases comprise inhibiting cancer growth, reducing leg swelling, symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, for reducing symptoms of pain; thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, comprising administering to a subject, in need thereof, an effective amount of the composition of this invention; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK In an embodiment the method is administering contacting an effective amount in a subject with the compound selected from formulas in this application.
A process and method for administration of the composition, wherein administration is by intravenous injection, intravenous drip, intraperitoneal injection or oral administration; wherein administration is by intravenous drip- 0.05-0 2mg/kg compound dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution, or by intravenous injection: 0.05-0.2mg/kg/day compound dissolved in 10-2OmI of 10% glucose solution or of 0.9% NaCI solution, or by intravenous drip: 0.1 -0.2mg/kg/day compound dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution, or by intravenous injection: 0.1 -0.2mg/kg/day compound dissolved in 1 0-2OmI of 10% glucose solution or of 0 9% NaCI solution, or by intraperitoneal injection(I.P ): 2 5mg/kg/day compound dissolved in 10% glucose solution or of 0.9% NaCI solution, or by oral administration wherein the dosage of mammal is 1 -10mg/Kg, 10-30mg/Kg, 30-60mg/Kg, or 60- 90mg/Kg compound, or by intravenous injection or intravenous drip wherein the dosage of mammal is 0.01 - 0.1 mg/Kg, 0.1 -0.2mg/Kg, 0.2 - 0.4mg/Kg, or 0.4 - 0.6 mg/Kg compound, or by intraperitoneal injection (LP.) wherein the dosage of mammal is 1 - 3mg/Kg, 3-5mg/Kg, 4-6mg/Kg, or 6-10mg/Kg compound.
The methods and uses of an isolated, purified or synthesized compound or its salt, ester, metabolite or derivative thereof, having the formula of :
named (1 A), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O- alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O- alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, and derivatives thereof; R4 represents CH2R6 or COR6, wherein R6 is selected from a group consisting of hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O-dibenzoyl, O-benzoyl, O- alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; R3 is H or OH; R8 is H or OH; R5 is a hydrogen, heterocyclic or sugar moiety(ies), wherein the sugar moiety(ies) is/are selected from a group consisting of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, and derivatives or combination thereof; wherein R9, R10, R1 1 , R12, R13, R14, R15 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; wherein at least two of R1 , R2 and R6 are compriseing a group selected from O-angeloyl, O-tigloyl, O- senecioyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; or at least one of R1 , R2, and R4 is a sugar moiety substituted with at least two groups selected from a group consisting of angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, aryl, acyl, heterocylic, heteroraryl, and a derivative thereof; orwherein R4 is CH2R6; wherein R1 and R2 independently consists an O-angeloyl group, or at least two of R1 , R2 and R6 are O-angeloyl or at least one of R1 , R2 or R6 is a sugar moiety with two O- angeloyls; or wherein R5 is/are the sugar moiety(ies) selected from the following sugars and alduronis acids: glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, glucuronic acid, galacturonic acid; or their derivatives thereof, or the combination thereof; wherein the sugar preferably comprises glucuronic acid, arabinose and galactose. In an embodiment, wherein R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof; in embodiment, the acyl has carbon 2 to 10. In an embodiment the method is administering contacting the compounds, wherein the compound is selected from the following: a) An isolated, purified or synthesized compound is having structure Xanifolia(Y),
or chemical name: 3-O-[β-D-galactopyranosyl (1 →2)]-α-l_-arabinofuranosy (1 →3)-β-D-glucuronopyranosyl-21 ,22-0-diangeloyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
b) An isolated, purified or synthesized compound is having structure Xanifolia (Y1 ),
or chemical name: 3-O-[β-D-galactopyranosyl (1 →2)]- α-L-arabinofuranosyl (1 →3)-β-D-glucuronopyranosyl-21 -0-(3,4-diangeloyl)-α-l_- rhamnophyranosyl-22-0-acetyl-3β,16α, 21 β, 22α, 28-pentahydroxyolean-12-ene;
c) An isolated, purified or synthesized compound is having structure Xanifolia (Y2),
or chemical name: 3-O-[β-D-glucopyranosyl- (1 →2)]-α-l_-arabinofuranosy (1 →3)-β-D-glucuronopyranosyl-21 ,22-0-diangeloyl-3β, 15α, 16α, 21 β, 22α, 24β, 28-heptahydroxyolean-12-ene;
d) An isolated, purified or synthesized compound is having structure Xanifolia (Y8), chemical name: 3-O-[/?-glucopyranosyl (1 →2)]- αr-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 , 22-0-diangeloyl-3/?, 16a, 2λβ, 22a, 24/?, 28-hexahydroxyolean-12-ene;
e) An isolated, purified or synthesized compound is having structure Xanifolia (Y9),
chemical name:3-0-[/?-galactopyranosyl (1 →2)]- α-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 -©-(S^-diangeloyl)-^- rhamnopyranosyl-28-0-acetyl-3/?, 16α, 2λβ, 22a, 28-pentahydroxyolean-12-ene; and
f) An isolated, purified or synthesized compound is having structure Xanifolia (Y10),
or chemical name:
3-0-[y5-galactopyranosyl (1 ^2)]-or-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 22-0-diangeloyl-3^, "[ 6a, 2\ β, 22a, 28-pentahydroxyolean-12-ene.
g) An isolated, purified or synthesized compound is having structure Xanifolia (YO),
or chemical name: 3-0-[β-D- galactopyranosyl(1 →2)]-α-l_-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl-21 -O- angeloyl, 22-0-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12- ene,
h) An isolated, purified or synthesized compound is having structure Xanifolia (X),
or chemical name: 3-0-{[/?-D-galactopyranosyl
(1 →2)]-[ol_-arabinofuranosyl (1 →3)]-/?-D-glucuronopyranoside butyl ester}-21 -0-acetyl- 22-0-angeloyl- 3#16tf,21#22#,28-pentahydroxyolean-12-ene.
i) An isolated, purified or synthesized compound is having structure (Y7),
or chemical name: 3-0-[jS-D-galactopyranosyl-
(1 →2)]-σ-L-arabinofuranosyl-(1 →3)-jS-D-glucuronopyranosyl-21 -O-angeloyl-28-0-2- methylbutanoyl-3/3, 15 a, 16σ, 21/3, 22σ, 28-hexahydroxyolean-12-ene
j) An
;
ACH-Y7; In an embodiment the method is administering contacting the compound, wherein the compound is selected from the following: k) An isolated, purified or synthesized compound is having a structure:
I) An isolated, purified or synthesized compound is having a structure:
In an embodiment the method is administering contacting the compound, wherein the compound is isolated, purified or synthesized having a structure selected from following formulas: wherein R1 , R2 are individually selected of an O-acetyl or O-angeloyl; wherein the R3,
R4, R5, R6, R7 is hydrogen or hydroxyl
In an embodiment the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the method is administering contacting the compound comprise of a triterpene wherein the carbon position 21 , 21 has an unsaturated group and sugar moieties at carbon 3.
In an embodiment, methods and compounds of this application reducing the adhesion ability inhibit bacteria in colonization and regulate tropism of cells.
In an embodiment, reducing the adhesion ability of cell or viruses in order to inhibit viruses binding to host cells, wherein the viruses comprise HIV.
This invention provides a use of compound for manufacture of medicament, a method and composition for modulating adhesion or anti-angiogenesis of cancer tumor, antiparasitics or manufacturing an adjuvant composition, wherein the modulating adhesion of cancer cell comprising modulating the secretion or expression of adhesion protein, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, cadherin, heparin, tenascin, CD 54, CAM; wherein the modulating comprises reducing, inhibiting and stimulating, wherein modulating adhesion protein comprises reducing the fibronectin for inhibiting the metastasis or growth of cancer cells, wherein the cancer is selected from breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, and renal cancer; wherein the modulating adhesion of cancer cell comprising modulating the secretion or expression of adhesion protein, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, cadherin, heparin, tenascin, CD 54, CAM; wherein the modulating comprises reducing, inhibiting and stimulating; wherein the modulating angiogenesis comprises inhibiting and stimulating angiopoietin, wherein comprising angiopoietini , angiopoietin 2, angiopoietin 3, angiopoietin 4, angiopoietin 5, angiopoietin 6 and angiopoietin 7; wherein the angiopoietin comprising angiopoietin-like 1 , angiopoietin-like 2, angiopoietin -like 3, angiopoietin-like 4, angiopoietin-like 5, angiopoietin-like 6 and angiopoietin-like 7; wherein the modulating comprises positive and negative regulating; wherein modulating angiopoietin comprises stimulating the angiopoietin 2 in order to inhibit angiogenesis; wherein modulating angiopoietin comprises inhibiting the angiopoietin 1 in order to inhibit angiogenesis; wherein modulating angiopoietin comprises inhibiting the angiopoietin-like 1 ; wherein modulating angiopoietin comprises inhibiting the angiopoietin-like 4; wherein the antiparasitics comprise inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria, wherein the method are is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the compound(s) are selected from ACH-Z4, ACH- Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5 and ACHMbI 2. In an embodiment the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, and Ba17. In an embodiment the saponins comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12, and Mb13.
In an embodiment, the methods and compositions of this application can be used for manufacturing an adjuvant vaccine, wherein the methods and compositions are used for manufacturing an adjuvant vaccine in a subject, wherein the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the compound(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5 and ACH-MbI 2. In an embodiment the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, and Ba 17. In an embodiment the saponins comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13. In an embodiment, this application provides an adjuvant composition comprising a saponin or compound selecting from above; wherein the composition is comprised of an immunostimulatory oligonucleotide.
In an embodiment, the methods and compositions of this application can be used for manufacture of medicament for vaccine or antiviral agent for Enterovirus comprising EV71 , wherein the method is administering contacting the cell or subject with compounds selected in this application comprising Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13.
The composition comprises the bioactive compounds from natural plants or synthesis. The majority of the plants are from the Sapindaceae family, which has 140-150 genera with 1400-2000 species. The program is based on our purification methods and biological assays including the MTT assay See International Application No. PCT/US05/31900, filed September 7, 2005, U.S. Serial No. 1 1 /289142, filed November 28, 2005, and U.S. Serial No. 1 1 /131551 , filed May 17, 2005, and PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008, the contents of which are incorporated herein by reference. The details of Analysis of gene expression of ES2 cells after Y-treatment by Microarray, Data Analysis Methods and Western blot in PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008, the contents of which are incorporated herein by reference
This invention provides a composition comprising an effective amount of triterpenoidal saponins named as Xanifolia Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, and YO or their salt or their derivatives. In an embodiment the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, and Ba17 or their salt or their derivatives. In an embodiment the saponins comprise Mb1 , Mb2, Mb3, Mb4, Mb5, Mb6, Mb7, Mb8, Mb9, Mb10, Mb1 1 , Mb12 and Mb13, or their salt or their derivatives for modulating the adhesion protein, reducing adhesion protein or reducing the secretion of fibronectin, for treating chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrheal, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of pain; for reducing symptoms of stomach pain; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, thrombosis, thromophlebitis; for treating rheumatism; for preventing gastric ulcers antispasmotic; blocking the migration, metastasis of cancer cells or inhibiting tumor growth. In an embodiment the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the compound(s) are selected from Compound Z1 to Z13 in the application. In an embodiment the compound(s) are selected from ACH-Z4, ACH- Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-Mb5 and ACH-MbI 2. The compounds of this invention can be isolated from natural sources or synthesized.
See experiments results in this application and see PCT/US05/31900, filed September 7, 2006; U.S. Serial No. 10/906,303, filed February 14, 2005; International Application No. PCT/US04/43465, filed December 23, 2004; International Application No. PCT/US04/33359, filed October 8, 2004 and U.S. Serial No. 1 1 /131551 , filed May 17, 2005, PCT/US2007/077273, filed August 30, 2007, PCT/US2008/002086, 1 188-ALA- PCT, filed February 15, 2008, US61 /038277 filed March 20, 2008, US61 /054308, filed May 19, 2008, the contents of which are incorporated herein by reference.
Acid Hydrolysis of Saponin
15 mg Xanifolia-Y was dissolved in 1 ml of Methanol. 1 ml of 2N HCI was then added. The mixture was refluxed in 8OC water bath for 5 hours. The solution was then neutralized by adding 2 ml of 1 N NaOH (to final pH 4-6). The aglycone was then extracted with ethylacetate 3 ml x 2. The extracts were collected and pooled. Further isolation of aglycone (ACH-Y) was achieved by HPLC with isocratic elution of 80 -100% acetonitrile. Repeat the experiment with compound Z4, Y10, Y2, Y8, Y7, YO, X, and ESCIN were obtained compounds ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E, ACH-MbI 2, ACH-Mb5.
In mild conditions, the saponin will be partial hydrolysis to a mixture of products. The procucts can be separated by HPLC. Also, specific partial hydrolysis can be achieved with enzymes. The β-glucosidase is good enzyme to cleave of the β-glucose from saponin.
Removal of the acyl group by alkaline hydrolysis
20 mg of Xanifolia-Y was dissolved in 0.5 ml of 1 M NaOH. The solution was incubated in 8OC water bath for 4 hours. It was cooled to room temperature before neutralized with 0.5 ml 1 N HCI (adjust pH to about 3). The mixture was extracted with 2 ml 1 -butanol 3 times. The butanol fractions were collected and lyophilized. The hydrolyzed saponin with further purified with HPLC in a C-18 column eluted with 25% acetonitrile.
Compounds AKOH-Y and AKOH-Mb5 have lost anticancer activity.
This invention provides a use of compound for manufacture of medicament or a method of modulating the adhesion proteins or their receptors, reducing the adhesion ability of the cancer cells, wherein the modulating comprises the positive or negative regulating. In an embodiment, the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, the method is reducing the secretion of fibronectin. This invention provides a method of blocking the migration, metastasis of cancer cells or inhibiting cancer cell growth or inhibiting leishmaniases modulating adhesion or inhibiting angiogenesis of cancer tumor, antiparasitics or manufacturing an adjuvant composition comprising administering an effective amount of a pharmaceutical composition comprising a composition comprises the molecular formula or compound in this invention. The cancers comprise Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma cancer, ovarian cancer, renal cancer, Prostate cancer, Breast cancer, bladder cancer, cervix cancer, liver cancer, bone cancer, brain cancer and Skin cancer. The compounds of this invention can be isolated from natural sources or synthesized. In an embodiment the method is administering contacting the compounds, wherein the compound is selected from the following:
(Z 1 ) 3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
(Z2) 3-0-[/3-D-galactopyranosyl-(1 →2)]-σ-L-arabinofuranosyl-(1 →3)-jS-D- -glucuronopyranosyl-21 -0-angeloyl-22-0-(angeloyl-2-methylbutanoyl) -3β, 15 α, 16α, 21/3, 22α, 28-hexahydroxyolean-12-ene;
(Z3) 3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -0-(2-methylpropanoyl), 22-0-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
(Z4) 3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0-benzoyl-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene;
(Z5) 3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0-angeloyl-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene;
(Z6) 3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -0-(2-methylpropanoyl)-0-benzoyl, 22-0-(2-methylpropanoyl)- 3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
(Z7) 3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -0-(2-methylpropanoyl)-0-angeloyl, 22-O-(2- methylbutanoyl) - 3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene; (Z8) 3-O-[β-D-galactopyraπosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21 -O-benzoyl, 22-O-benzoyl-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene;
(Z9) 3-O-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0-benzoyl-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene;
0 3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl -(1 →3)-β-D-glucuronopyranosyl- 21 -0-(2-methylpropanoyl), 22-0-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene;
3-0-[/3-D-galactopyranosyl-(1 →2)]- β- D-xylopyranosyl -(1 →3)-j8-D- -glucuronopyranosyl-21 -0-angeloyl-22-0-(angeloyl-2-methylbutanoyl) -3/3, 15 a, 16σ, 21/3, 22α, 28-hexahydroxyolean-12-ene;
2,
3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-angeloyl, 22-0- tigloyl -3β, 16α, 21 β, 22α, 28-pentahydroxyolean-12-ene;
3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl- 21 -O-angeloyl, 22-0- tigloyl -3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
This invention provides uses of a compound for manufacture of medicament selected from formula (1 B), for modulating, regulating or interact with the adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, this invention provides a method of reducing the secretion of fibronectin, modulating adhesion or angiogenesis of cancer cells, antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, inhibiting cancer metastasis or growth, using the compounds selected for the following:
, also named as (1 B), or a salt, ester, metabolite or derivative thereof, wherein R1 comprises a group selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, dibenzoyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, acyl, aryl, heterocylic, heteroraryl, alkenylcarbonyl and derivatives thereof; R2 comprises a group selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkenylcarbonyl and derivative thereof; R4 represents CH2OR6 or COOR6, wherein R6 is selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and derivative thereof; R3 is H or OH; wherein at least one of R1 , R2,and R6 comprises a group selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and derivative thereof; R5 comprises a hydrogen or sugar moiety, wherein the sugar moiety comprises at least one sugar of, but is not limited to, D-glucose, D-galactose, L- rhamnose, L-arabinose, D-xylose, alduronic acid: D-glucuronic acid, D-galacturonic acid or a derivative thereof, or the combination thereof.
In an embodiment, R1 comprises a sugar moiety wherein substituted with two groups selecting from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic heteroraryl and a derivative thereof. In an embodiment, R1 comprises a sugar moiety wherein substituted with at least one group selecting from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof. In an embodiment, R2 comprises a sugar moiety wherein at least one group is selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof. In an embodiment, R2 comprises a sugar moiety or a side chain wherein at least two groups are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof. In an embodiment, R4 comprises CH2OR6 or COOR6 wherein R6 is a sugar moiety which comprises at least one group selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof. In an embodiment, R4 comprises CH2OR6 orCOOR6, wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substitutedalkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof. In an embodiment, R4 comprises CH2OR6 or COOR6, wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl and senecioyl. In an embodiment, R4 comprises CH2OR6 or COOR6 of formula (1 B), at least two of R1 , R2 and R6 comprise the group selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl and a derivative thereof. In an embodiment, R4 comprises CH2OR6 or COOR6 of formula (1 B), wherein at least two of R1 , R2 and R6 comprise angeloyl, benzoyl, alkenoyl, or a derivative thereof. In an embodiment, R4 is a side chain comprising CH2OCOCH3, CH2COO-alkyl, CH2OH, COOH, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, or a derivative thereof. In a further embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises one or more sugar of, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, or alduronic acid: glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof. In an embodiment, R5 comprises a sugar moiety or a group capable of performing the function of the sugar moiety. In an embodiment, the R5 represents H. In an embodiment, R4 represents H, OH or CH3. In an embodiment, position C23, C24, C25, C26, C29 and C30 of the compound independently comprise CH3, CH2OH, CHO, COOH, COOa-lkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, acetyl group or derivatives thereof, particular CH3. In an embodiment, R1 and R2 independently comprise an angeloyl group. In an embodiment, R1 is a sugar moiety or a side chain which comprise two angeloyl groups. In an embodiment, R1 and R2 independently comprise a benzoyl group. In an embodiment, R1 is a sugar moiety which is substituted with two benzoly groups. In an embodiment, R3 represents H or OH. In an embodiment, R8 may be OH. In an embodiment, the O at C21 , 22 may be replaced by N. In an embodiment, this invention provides a method of reducing the secretion of fibronectin; wherein the medicament is for inhibiting tumor or cancer cell growth and for treating cancer, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer or cervix cancer. Substitution, deletion and/or addition of any group in the above- described compounds by other group(s) will be apparent to one of ordinary skill in the art based on the teachings of this application. In a further embodiment, the substitution, deletion and/or addition of the group(s) in the compound of the invention does not substantially affect the biological function of the compound.
This invention provides uses, methods, processes, compounds and compositions for modulating adhesion or angiogenesis of cancer cells, antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, inhibiting cancer metastasis or growth, reducing adhesion protein of cells, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, methods comprise inhibiting the gene expression. In an embodiment, this invention provides a method of reducing the secretion of fibronectin. In an embodiment the method can block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti-angiogenesis, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer, cervix cancer, esophagus cancer, testis cancer, spleen cancer, kidney cancer, lymph cancer, pancreas cancer, stomach cancer and thyroid cancer, In an embodiment the compounds are anti-angiogenesis, inhibiting cancer cell metastasis and inhibiting cancer growth, In an embodiment the compounds promote angiopoietin 2 . In an embodiment the compound is selected from the following formulas (1 E). In an embodiment the method is administering contacting the compounds, wherein the compound is selected from the formula (1 E):
, also named (1 E), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, 0-senecιoyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R4 represents CH3, CHO, CH2R6 or COR6, wherein R6 is selected from hydroxyl, O- angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O- alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O- alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, O- alkenylcarbonyl and derivatives thereof; R3 is H or OH; R8 is H or OH, particularly OH, R16 is H ,or R4 and R16 may together form -CH2-X-, CH(OH)-X- or C(=O)-X-, wherein the -X- may be O or N ; wherein when the C12-13 of ring 3 of the triterpene has a double bond then R16 is absent.
R5 is a hydrogen, heterocyclic or sugar moiety(ies), wherein the sugar moiety(ies) is/are selected from a group consisting of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, and derivatives or combination thereof; wherein R9, R10, R1 1 , R12, R13, R14, R15 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; wherein R4 and R16 form a divalent radical of formula CH2O, CH(OR7)O, or COOR7, wherein R7 is hydrogen, alkyl, angeloyl, tigloyl, senecioyl, dibenzoyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, and derivatives thereof; wherein at least two of R1 , R2 and R6 are compriseing a group selected from O-angeloyl, O-tigloyl, O-senecioyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O- benzoyl alkyl substituted O-alkanoyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; or at least one of R1 , R2, and R4 is a sugar moiety substituted with at least two groups selected from a group consisting of angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, and a derivative thereof; or wherein R4 is CH2R6; wherein R1 and R2 independently consists an O-angeloyl group, or at least two of R1 , R2 and R6 are O-angeloyl or at least one of R1 , R2 or R6 is a sugar moiety with two O- angeloyls; wherein R5 is/are the sugar moiety(ies) selected from the following sugars and alduronis acids: glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, glucuronic acid, galacturonic acid; or their derivatives thereof, or the combination thereof; wherein the sugar preferably comprises glucuronic acid, arabinose and galactose. In an embodiment, wherein R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof. In an embodiment, wherein R5 is 3-β-O-{[(α-L-rhamnopyranosyl-(1 →2)]-α-L-rhamnopyranosyl--(1 →2)-β-D- galactopyranosyl--(1 →3)]-[β-D- galactopyranosyl--(1 →2)]- β-D-glucuronopyranosyl}. In an embodiment, wherein the carbon ring 3 comprises a double bond when R16 is H; wherein the double bond in carbon ring 3 is reduced when R4 and R16 form a divalent radical.
In an embodiment the method is administering contacting the compounds, wherein the compound is selected from the formula (1 F): , also named (1 F), wherein
R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R4 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R3 is H or OH; R8 is H or OH, particularly OH; R5 is a hydrogen or sugar moiety(ies), wherein the sugar moiety(ies) is/are selected from a group consisting of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, and derivatives or combination thereof; wherein R9, R10, R1 1 , R12, R13, R14, R15 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; wherein at least two of R1 , R2 and R4 are compriseing a group selected from O-angeloyl, O-tigloyl, O-senecioyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; or at least one of R1 , R2, and R4 is a sugar moiety substituted with at least two groups selected from a group consisting of angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, and a derivative thereof; or wherein R4, R1 and R2 independently consists an O-angeloyl group, or at least two of R1 , R2 and R4 are O-angeloyl or at least one of R1 , R2 or R4 is a sugar moiety with two O-angeloyls; wherein R5 is/are the sugar moiety(ies) selected from the following sugars and alduronis acids: glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, glucuronic acid, galacturonic acid; or their derivatives thereof, or the combination thereof; wherein the sugar preferably comprises glucuronic acid, arabinose and galactose. In an embodiment,wherein R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof; In an embodiment, wherein R5 is 3-βO-{[(α-L-rhamnopyranosyl-(1 →2)]-α-L- rhamnopyranosyl-(1 →2)-β-D- galactopyranosyl--(1 →3)]-[β-D- galactopyranosyl- (1 →2)]- β-D-glucuronopyranosyl}
In an embodiment the method is administering contacting the compounds of following:
also named (1 G), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R4 represents CH3, CHO, CH2R6 or COR6, wherein R6 is selected from hydroxyl, O- angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O- alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O- alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, O- alkenylcarbonyl and derivatives thereof; R3 is H or OH; R5 is H or OH; wherein R6, R7, R8, R9, R10, R1 1 , R12 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO- heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; In an embodiment the method is administering contacting the compounds of following:
also named (1 H), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R4 is selected from hydroxyl, CH2OH, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof;
R3 is H or OH; R5 is H or OH; wherein R6, R7, R8, R9, R10, R1 1 , R12 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO-alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; In an embodiment the use or method is administering contacting the compounds of following:
named (U), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O- dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O- heterocylic, O-heteroraryl, O-alkenylcarbonyl and derivatives thereof; R4 represents CH3, CHO, CH2R6 or COR6, wherein R6 is selected from hydroxyl, O- angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O- alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O- alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, O- alkenylcarbonyl and derivatives thereof; R3 is H or OH ; R5 is H or OH, particularly OH; wherein R6, R7, R8, R9, R1 0, R1 1 , R12 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO- alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; R1 3 is COOH or COO-alkyl,
; also named (1 L) , wherein R1 , R2 are angeloyl, R3 is OH, R4 is CH2OH, R5 is H or OH
In an embodiment the method is administering contacting the compounds of following:
or 3-0-[β-D-galactopyranosyl(1 →2)] -β- D-xyopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-angeloyl, 22-O-angeloyl-3β, 15a, 16a, 21 β, 22a, 28-hexahydroxyolean-12-ene,
3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xyopyranosyl (1 →3)-β-D-glucuronopyranosyl-21 - O-benzoyl, 22-0-benzoyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene
3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xyopyranosyl (1 →3)-β-D-glucuronopyranosyl-21 - 0-(2-methylpropanoyl)-0-angeloyl, 22-O-(2- methylbutanoyl) -3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene,
A sugar moiety is a segment of a molecule comprising one or more sugar groups. Substitution, deletion and/or addition of any group in the above-described compounds will be apparent to one of ordinary skill in the art based on the teaching of this application. In a further embodiment, the substitution, deletion and/or addition of the group(s) in the compound of the invention does not substantially affect the biological function of the compound.
A method or a use of compound for manufacture of medicament of inhibiting venous insufficiency, particularly hemorrhoids or inhibiting leg swelling, or peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, Expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrheal, hemorrhoids, episiotomies, hamonhoids, peripheral edema formation or postoperative swelling; for reducing symptoms of pain; for reducing symptoms of stomach pain; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic comprising administering to a subject, in need thereof, an effective amount of the composition of any one of the above compounds or a compound comprises a triterpene which comprises any two of angeloyl, tigloyl, senecioyl, perferable two angeloyl groups, and a sugar moiety, glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid or galacturonic acid, or a derivative thereof, or the combination thereof, preferable selected from glucuronic acid, galacturonic acid, glucose, galactose and arabinose. The method is regulating or interacting with adhesion protien, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK. In an embodiment, the method is reducing the secretion of fibronectin. In an embodiment, the method comprises inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition. In an embodiment, antiparasitics comprise inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria.
This invention provides a method for inhibiting the growth, migration, metastasis of cancer by altering the characteristic of membrane of cancer cell, wherein the characteristic comprise reducing adhesion protein; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK; wherein comprising inhibiting the secretion of fibronectin, wherein comprising administering to a subject, in need thereof, an appropriate amount of triterpenoidal saponins comprising two or more angeloyl groups, or a compound comprises a triterpene which comprises any two of angeloyl, tigloyl, senecioyl, perferable two angeloyl groups, and a sugar moiety, glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid or galacturonic acid, or a derivative thereof, or the combination thereof, preferably selected from glucuronic acid, galacturonic acid, glucose, galactose and arabinose. This invention provides a composition comprising an effective amount of the compound of any one of compound selected from the above formula or a salt, ester, metabolite or derivative thereof as a medicament for reducing adhesion protein; wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK, for inhibiting the growth, migration, metastasis of cancer, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer or cervix cancer.
This invention also provides a composition comprising the above described compounds or their derivatives for reducing adhesion protein, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, Glycosylation cell surface proteins, polyglycans, cadherin, heparin, tenascin, CD 54, CAM, elastin and FAK; wherein comprising inhibiting the secretion of fibronectin, wherein for treating venous insufficiency, particularly hemorrhoids or inhibiting leg swelling, or peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, Expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, episiotomies, hemonhoids, peripheral edema formation or postoperative swelling; for reducing symptoms of pain; for reducing symptoms of stomach pain; for reducing symptoms of leg pain; for treating pruhtis, lower leg volume, thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, inhibiting leishmaniases , for modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition. In an embodiment of the above, the uses of compositions comprising any one of triterpenoid saponins with the following formula: 3-0-{[/?-D-galactopyranosyl (1 →2)]-[αr-l_-arabinofuranosyl (1 →3)]-/?-D- glucuronopyranoside butyl ester}-21 -0-acetyl-22-0-angeloyl-3/?,16α;21/?,22α,28- pentahydroxyolean-12-ene.
3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl- 21 ,22-O-diangeloyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene, 3-0-[β-D-galactopyranosyl(1 →2)]-α-l_-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl - 21 -0-(3,4-diangeloyl)-α-L-rhamnophyranosyl-22-0-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene,
3-0-[β-D-glucopyranosyl-(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl- 21 ,22-0-diangeloyl-3β, 15α, 16α, 21 β, 22α, 24β, 28-heptahydroxyolean-12-ene, 3-0-[/?-glucopyranosyl (1 →2)]-α-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 , 22- O-diangeloyl-3/?, 16«; 21$ 22a, 2Λβ, 28-hexahydroxyolean-12-ene, 3-0-[/?-galactopyranosyl (1 — >2)]-cr-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 -O- (3,4-diangeloyl)-α-rhamnopyranosyl-28-0-acetyl-3^, 16a, 21 β, 22a, 28- pentahydroxyolean-12-ene,
3-0-[/?-galactopyranosyl (1 →2)]-ctr-arabinofuranosyl (1 →3)-^-glucuronopyranosyl-21 , 22-0-diangeloyl-3/?, 16a, 2*1 /3, 22a, 28-pentahydroxyolean-12-ene,
3-0-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosy I- 21 -O-angeloyl, 22-0-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene,
3-0-[/3-D-galactopyranosyl-(1 →2)]-σ-L-arabinofuranosyl-(1 →3)-/3-D- -glucuronopyranosyl-21 -0-angeloyl-28-0-2-methylbutanoyl-3)8, 15 a, 16a, 21 )8, 22σ, 28- hexahydroxyolean-12-ene
This invention also provides a composition for regulating or reducing adhesion protein, wherein the adhesion protein comprising fibronectin, integrins family, CD44, Myosin Vl, vitronectin collagen, laminin, Glycosylation cell surface proteins, polyglycans and FAK; inhibiting venous insufficiency, particularly hemorrhoids or inhibition of leg swelling, or inhibiting cancer growth, inhibiting leishmaniases, modulating adhesion of cancer cells, modulating angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition, comprising any of compounds selected from the following compounds (A) to (X) and (A1 ) to (X1 ) incorporated here from PCT/US2008/002086, 1 188-ALA-PCT, :
In an embodiment, a triterpene comprise the following structure has activities of reducing adhesion proteins to blocks the migration, metastasis of cancer cells, growth of cancers, inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics, or manufacturing an adjuvant composition.
wherein at least two of R1 , R2 and R3 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof. In an embodiment, at least one of R1 , R2 and R3 comprise a sugar moiety comprising two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof. In embodiment, R1 , R2 or R3 comprise angeloyl groups, tigloyl groups, senecioyl groups or acetyl group or their combinations, preferable wherein at least two of the R1 , R2 and R3 comprise angeloyl groups. In an embodiment, R5 comprises sugar moiety. In an embodiment, the sugar moiety comprises at least one sugar, or glucose, or galactose, or rhamnose, or arabinose, or xylose, or alduronic acid, or glucuronic acid, or galacturonic acid, or their derivative thereof, or the combination thereof. In an embodiment, the sugar moiety comprises one or more sugar selected from, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof. In an embodiment, the sugar moiety comprise glucose, galactose or arabinose, or combination thereof, or derivatives thereof. In an embodiment, the sugar moietiy comprise alduronic acids, galactose and arabinose, wherein the alduronic comprise glucuronic acid or galacturonic acid. In an embodiment, the sugar moiety comprise alduronic acids, glucose and arabinose, wherein the alduronic comprise glucuronic acid or galacturonic acid. In an embodiment, R5 is Hydrogen. In an embodiment, the R1 , R 2 and R3 may be attached in other position of the structure.
In an embodiment, the compound is thterpenoid saponin comprise comprises at least two angeloyl groups, tigloyl groups, senecioyl groups or acetyl group or their combinations, preferable wherein at least two angeloyl groups.
In an embodiment, at least two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof.
In an embodiment, at least one of the side bonds comprise a sugar moiety comprising two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, alkanoyl substituted phenyl, alkenoyl substituted phenyl, alkenylcarbonyl or substituted with an C2-9 acid or derivative thereof. In an embodiment, the compound comprises a sugar moiety. In a further embodiment, the sugar moiety comprises glucose, galactose or arabinose or combination thereof.
In a further embodiment, the sugar moiety comprises at least one sugar, or glucose, or galactose, or rhamnose, or arabinose, or xylose, or alduronic acid, or glucuronic acid, or galacturonic acid, or their derivative thereof, or the combination thereof.
In a further embodiment, the sugar moiety comprises one or more sugar selected from, but is not limited to glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, or derivatives thereof, or the combination thereof.
A composition comprising an effective amount of compound selected from the above formula or a salt, ester, metabolite or derivative thereof as a medicament for regulating or reducing adhesion protein, blocking the migration, metastasis of cancer cells, inhibiting tumor or cancer cell growth and for treating cancer, wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal and cervix cancer.
In a further embodiment, a compound or sapongenin comprises the structure (d) or (e) has anti-cancer or inhibiting virus activities.
A composition for regulating or reducing adhesion protein, blocking the migration, metastasis of cancer cells, treating cancers or inhibiting virus, comprising a compound, wherein the compound is a triterpene, which comprises at least two side chains which comprise angeloyl groups, wherein the side chains are at adjacent carbon in trans position. In an embodiment, the side chains are at alternate carbon in cis position. In an embodiment, the side chains are at alternate carbon in trans position. In an embodiment, the side chains are attached an acyl. In an embodiment, the side chains are attached an unsaturated group.
In an embodiment, the side chains are in non-adjacent carbon cis or trans position. In an embodiment, the side chains comprise a functional group capable of performing the function of angeloyl group. The above compounds can be used for regulating or reducing adhesion protein, blocking the migration, metastasis of cancer cells, inhibiting tumor cell growth, reducing leg swelling, symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, expectorant, peripheral vascular disorders, by administering to a subject in need thereof, an effective amount of the above described compounds.
This invention provides a method for inhibiting tumor cell growth, regulating cell growth, reducing inflammation, inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics or manufacturing an adjuvant composition, comprising administering to a subject, in need thereof, an effective amount of the compound which comprises any of the above structures to said subject. The cancers are included but not limited to Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma cancer, Ovarian cancer, Renal cancer, Prostate cancer, Breast cancer, bladder cancer, cervix cancer, liver cancer, bone cancer, brain cancer and Skin cancer.
This invention also provides a method for reducing swelling, reducing symptoms of chronic venous insufficiency, peripheral edema, antilipemic, chronic venous disease, varicose vein disease, varicose syndrome, venous stasis, Expectorant, peripheral vascular disorders, cerebro-organic convulsion, cerebral circulation disorder, cerebral edema, psychoses, dysmenorrhea!, hemorrhoids, episiotomies, peripheral edema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, for reducing symptoms of pain; thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, comprising administering to a subject, in need thereof, an effective amount of the composition of this invention.
This invention provides a composition comprising the compounds provided in the invention for treating cancers; for inhibiting virus; for preventing cerebral aging; for improving memory; improving cerebral functions, for curing enuresis, frequent micturition, urinary incontinence, dementia, Alzheimer's disease, autism, brain trauma, Parkinson's disease or other diseases caused by cerebral dysfunctions; for treating arthritis, rheumatism, poor circulation, arteriosclerosis, Raynaud's syndrome, angina pectoris, cardiac disorder, coronary heart disease, headache, dizziness, kidney disorder; cerebrovascular diseasea; inhibiting NF-Kappa B activation; for treating brain edema, sever acute respiratory syndrome, respiratory viral diseases, chronic venous insufficiency, hypertension, chronic venous disease, anti-oedematous, anti inflammatory, hemonhoids, peripheral edema formation, varicose vein disease, flu, post traumatic edema and postoperative swelling;for inhibiting blood clot, for inhibiting ethanol absorption; for lowering blood sugar; for regulating the adrenocorticotropin and corticosterone level. This invention provides a composition for AntiMS, antianeurysm, antiasthmatic, antibradykinic, anticapillarihemorrhagic, anticephalagic, anticervicobrachialgic, antieclamptic, antiedemic, antiencaphalitic, antiepiglottitic, antiexudative, antiflu, antifracture, antigingivitic, antihematomic, antiherpetic, antihistaminic, antihydrathritic, antimeningitic, antioxidant, antiperiodontic, antiphlebitic, antipruritic, antiraucedo, antirhinitic, antitonsilitic, antiulcer, antivaricose, antivertiginous, cancerostatic, corticosterogenic, diuretic, fungicide, hemolytic, hyaluronidase inhibitor, lymphagogue, natriuretic, pesticide, pituitary stimulant, thymolytic, vasoprotective, inhibiting leishmaniases, modulating adhesion or angiogenesis of cancer cells, antiparasitics, or manufacturing an adjuvant composition and venotonic treatment.
Alkenyl means unsaturated linear or branched structures and combinations thereof, having 1 -7 carbon atoms, one or more double bonds therein. Non-limiting examples of alkenyl groups include vinyl, propenyl, isopropenyl, butenyl, s- and t-butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, and hexadienyl.
An aryl is a functional group of organic molecule derived from an aromatic compound such as benzene, a 6-14 membered carbocyclic aromatic ring system cpmprising 1 -3 benzene rings. If two or more aromatic rings are present, then the rings are fused together, so that adjacent rings share a common bond. Examples include phenyl and naphthyl. The aryl group may be substituted with one or more sunstitutes independnetly selected from halogen, alkyl or alkoxy.
Acyl is a functional group obtained from an organic acid by the removal of the carboxyl. Acyl groups can be written as having the general formula -COR, where there is a double bond between the carbon and oxygen. The names of acyl groups typically end in -yl, such as formyl, acetyl, propionyl, butyryl and benzoyl.
Benzoyl is one of acyls, C6H5COR, obtained from benzoic acid by the removal of the carboxyl.
Heterocyclic compound - a compound containing a heterocyclic ring which refers to a non-aromatic ring having 1 -4 heteroatoms said ring being isolated or fused to a second ring selected from 3- to 7-membered alicyclic ring containing 0-4 heteroatoms, aryl and heteroaryl , wherein said heterocyclic comprises pyrrolidinyl , pipyrazinyl , morpholinyl , trahydrofuranyl, imidazolinyl, thiomorpholinyl , and the like.
Heterocyclyl groups derived from heteroarenes by removal of a hydrogen atom from any ring atom.
Alkanoyl is the general name for an organic functional group RCO-, where R represents hydrogen or an alkyl group. Preferably alkanoyl is selected from acetyl, propionoyl, butyryl, isobutyryl, pentanoyl and hexanoyl.
Alkenoyl is alkenylcarbonyl in which alkenyl is defined above. Examples are pentenoyl(tigloyl) and hexenoyl(angeloyl).
Alkyl is a radical containing only carbon and hydrogen atoms arranged in a chain, branched, cyclic or bicyclic structure or their combinations, having 1 -18 carbon atoms. Examples include but are not limited to methyl, ethyl, propyl isopropyl, butyl, s- and t- butyl, pentyl, hexyl, heptyl, octyl, nonyl, undecyl, dodecyl, tridecyl, tetradecyl, pentadecyl, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
Benzoyl alkyl substituted alkanoyl is refer to straight or branched C1 -C6 alkanoyl substituted with at least one benzoyl and at least one alkyl, wherein the benzoyl is attached to a straight or branched C1 -C6 alkyl. Preferably a benzoyl alkyl substituted alkanoyl is benzoyl methyl isobutanoyl.
A sugar moiety is a segment of molecule comprising one or more sugars or derivatives thereof or alduronic acid thereof.
Isobutyryl is Synonym of 2-Methylpropanoyl Y and Y3 represent the same compound. YM and (ACH-Y) represent the same compound. (Z)-O(C=O)C(CH3)=CH-C6H5 is [O- C5 alkenoyl substituted with phenyl]
(Z)-O(C=O)CH=CH-C6H5 is [0-C4 alkenoyl substituted with phenyl]
(E)-O(C=O)CH=CH-C6H5 is [0-C4 alkenoyl substituted with phenyl]
O(C=O)C6H5, Benzoyl is [0-C2 alkanoyl substituted with phenyl]
(2Z)-2-methyl-3-phenylacrylic acid
(2Z)-3-phenylacrylic acid
(2E)-3-phenylacrylic acid
Benzoic acid
O-Angeloyl: O(C=O)C(CH3)=CH(CH3) This invention provides a method of altering the characteristic of cancer cell membrane to block the migration, metastasis of cancer cells or inhibit the growth of cancers or anti- angiogenesis.
This invention provides a composition and method for inhibiting the growth, migration, metastasis of cancer by altering the adhesion characteristic of membrane of cancer cell, wherein the cancers comprise breast cancer, leukocyte cancer, liver cancer, ovarian cancer, bladder cancer, prostate cancer, skin cancer, bone cancer, brain cancer, leukemia cancer, lung cancer, colon cancer, CNS cancer, melanoma cancer, renal cancer or cervix cancer, wherein the method is administering contacting Xanifolia YO, Y1 , Y2, Y, Y5, Y7, Y8, Y9, Y10, or a salt, ester, metabolite thereof. In an embodiment the method is administering contacting the compound selected from formula in this application.
This application shows Xanifolia-Y is an alternate or supplemental agent to DNA- inhibition or microtubule-targeting drugs. It could be beneficial if it is used singly or in combination with other drugs of different mechanisms (block M-phase progression or DNA synthesis). Our inventions show combined effect of Xanifolia-Y and paclitaxel on inhibition of ES2 cells' growth (Detail in Experiment 14 U.S. Serial Nos.1 1/683198, filed on March 7, 2007) .
Identify the binding target of Xanifolia-Y of adhesion proteins and signaling proteins in ovarian cancer cells.
In our animal studies, it was shown that Xanifolia-Y extended the life span of tumor bearing mice. (Reference Experiments 7, 8, 9 in U.S. Serial Nos.1 1/683198, filed on March 7, 2007,). The animals died sooner if the treatment of Xanifolia-Y was delayed (comparing results of treatments started from 1 , 4 or 10 days after tumor inoculation). The results show that Xanifolia-Y inhibits migration or metastasis of the inoculated cancer cells. Ovarian carcinoma cells express high levels of adhesion molecules. Adhesion proteins are present in both cancer cells and mesothelial cells. While the lost of adhesion blocks of the protein accessibility due to a result of modulating by Xanifolia- Y, In an embodiment, the interaction of Xanifolia-Y with membrane alter the adhesion protein's binding site(s). Fibronectin is a kind of glycoprotein that binds to membrane spanning receptor proteins comprising the integrins, collagen, fibrin and heparin sulfate. Fibronectin has been implicated in tumor development and metastasis. This application provides methods and compositions for modulating the gene expression of fibronectin, inhibiting the secretion of fibronectin, reducing the receptors of fibronectin, reducing the adhesion ability fibronectin, inhibiting the metastasis, or inhibiting cancer growth, wherein the method and composition comprises administering to the said subject as effective amount of compounds selected in this appliaction.
Angiogenesis is a process involving the growth of new blood vessels. It is a normal process in growth and development. However, this is also a fundamental step in the transition of tumors from a dormant state to a malignant state. The angiopoietins are protein growth factors that modulate angiogenesis. The identified angiopoietins comprise angiopoietin 1 , angiopoietin 2, angiopoietin 3, angiopoietin 4, angiopoietin 5, angiopoietin 6, angiopoietin 7, angiopoietin-like 1 , angiopoietin-like 2, angiopoietin-like 3, angiopoietin-like 4, angiopoietin-like 5, angiopoietin-like 6, and angiopoietin-like 7. In an embodiment, the angiopoietin 1 is a positive foctor to promote the new blood vessels. In embodiment, the angiopoietin 2 is antagonist of angiopoietin 1 , which is a negative factor for the growth of new blood vessels. This application provides methods and compositions for modulating angiopoietin and inhibiting cancer growth; wherein the cancers comprise breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal and cervix cancer, wherein the methods and compositions comprise administering to the said subject as effective amount of compounds selected in this appliaction. The compounds in this application are positive regulating angiopoietin 2. The compounds in this application are negative regulating the angiopoietin 1 . The results of the micro array experiment showed that compound Y and YM (ACH-Y) modulate the gene expression of angiopoietin family in ES2 cells. They promote angiopoietin 2 and inhibit angiopoietin 1 and angiopoietin-like 1 and angiopoietin-like 4.
The compounds in this application are used antiparasitics, enhancing an immune response, providing adjuvant activities or providing vaccine activities, anti-angiogenesis, inhibiting cancer cell metastasis and inhibiting cancer growth, wherein the compounds comprise Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, ACH-Y or a salt, ester, metabolite thereof and compounds selected from formula (1 A), (1 B), (1 C) , (1 D), (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment the method is administering contacting the compound in this application comprising Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, Xanifolia (x), Escin or Aescin or a salt, ester, metabolite thereof. In an embodiment the compound may be selected from formulas (1 A), (1 B), (1 C), (1 D) (1 E), (1 F), (1 G), (1 H), (U), (1 K), (1 L). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application. In an embodiment the compound(s) are selected from Compound Z1 to Z7 in the application. In an embodiment the compound(s) are selected from ACH-Z4, ACH-Y10, ACH-Y2, ACH-Y8, ACH-Y7, ACH-YO, ACH-X, ACH-E. In an embodiment the saponins comprise Ba1 , Ba2, Ba3, Ba4, Ba5, Ba6, Ba7, Ba8, Ba9, Ba10, Ba1 1 , Ba12, Ba13, Ba14, Ba15, Ba16, Ba 17.
The triterpene compounds of this invention can be administered to a subject in need thereof treating the subject, wherein including preventing canceror has adjuvant effect to the subject, or inhibiting the initation or promotion of cancer, or killing the cancer/tumor cells. In an embodiment the compounds inhibit the activation of nuclear factor-kB, wherein inhibiting the localization or wherein binding the DNA. In an embodiment the compounds induce apoptosis in cancer cells. The triterpene compounds of this invention can reduce blood vessel in the tumor in a subject. (Figure 1 )
Table 1 to 12, Effect of Y and YM on gene expression (Table of 1 to 12 PCT/US2008/002086, 1188- AL A- P CT, filed February 15, 2008 are incorporated herein by reference )
Table 13: Effects of Y and YM on Glypican expression in ES2 cells
The results of the micro array experiment showed that compound Y and Ym inhibit gene expression of Glypican in ES2 cells.
Table 14: Effects of Y and YM on regulator of G-protein expression in ES2 cells
The results of the micro array experiment showed that compound Y and YM inhibit gene expression of G-protein in ES2 cells
Table 15: Effects of Y and YM on thrombospondin in ES2 cells
The results of the micro array experiment showed that compound Y and YM inhibit gene expression of thrombospondin in ES2 cells
Table 16: Effects of Y and YM on insulin-like growth factor binding protein expression in ES2 cells
The results of the micro array experiment showed that compound Y and YM inhibit gene expression of insulin-like growth factor binding protein in ES2 cells.
Table 17: Effects of Y and YM on RAB3B, member RAS oncogene family protein expression in ES2 cells
The results of the micro array experiment showed that compound Y and Ym inhibit gene expression of RAB3B, member RAS oncogene family protein in ES2 cells. Table 18: Effects of Y and YM on potassium channel, subfamily U, protein expression in ES2 cells
The results of the micro array experiment showed that compound Y and Ym inhibit gene expression of family protein relate to potassium channel in ES2 cells.
Fibronectin secretion studies summary:
Reduction of Fibronectin secretion from ES2 cells after xanifolia-Y treatment. (Results of F1 , F3, F4, F5, F7, F8, F1 1 , F12A, F15, F16, F17) Data in
PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008 are incorporated herein by reference
(F1 1 ) For lung carcinoma cells (H460), at concentration of 20ug/ml, there are inhibitions of Fibronectin secretion ranged from 20-60%. (F12A) For bladder carcinoma cells (HTB-9), Xanifolia-Y (10 ug/ml) inhibits 50% of
Fibronectin secretion.
(F15) In liver HepG2 cells. 10 ug/ml of xanifolia-Y inhibits 42% secretion of Fibronectin. (F16) Incubation of brain glioblastoma T98G cells with10ug/ml of xanifolia-Y inhibits 27% Fibronectin secretion and with 20ug/ml Y inhibits74% Fibronectin secretion. (F17) For skin SK-Mel-5 cells, the inhibition is 40-57% with 20 ug/ml of Xanifolia-Y.
Studies of xanifolia-Y analogs and other saponin on Fibronectin secretion from ES2 cells. Results of (F 23, F21 , F 13, F14, F24, F25, 26, 31 B, F27,29, F28, 30, F 31 , 32, F 33, F20) in PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008)
Studies of other saponins on Fibronectin secretion from ES2 and other cells
Liver
Lung
Bladder
Brain
Skin
Up regulation of Angiopoietin 2 (Ang2) in ES2 cells with Xanifolia-Y treatment.
The results of study in PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008 are incorporated herein by reference.
Data obtained from our Microarray experiments disclose that Xanifolia Y modulates gene expression of the following genes (represented by gene symbol):
Gene Symbol: ABL2, ADAMTS1 , AKR1 C3, AMIGO2, ANGPT2, ANKRD1 1 , AP2B1 ,
APEH, APLP2, ARL10C, ARMC4, ARMCX1 , ARMCX6, ARNTL2, ARNTL2, ATF3,
ATP6V0E, ATP6V1 B2, ATP6V1 C1 , ATP6V1 C1 , BCL2A1 , BCL6, BRI3, BTD, C14orf109, C14orf78, C17orf32, C6orf65,C9orf10, C9orf103, CAD, CAV1 , CAV2, CBLL1, CCL20, CD33L3, CEBPB, CEP4, CFH /// CFHL1, CHRDL1, CITED2, CITED2, CLDN14, CLN8, CLTA, CNAP1, COG6, COL18A1, COL4A2, COL5A1, COL5A2, COL6A3, COPG, CPM, CPNE3, CPSF1, CSRP2BP, CSTB, CTNS, CXCL2, DDB1, DDIT3, DDX20, DKFZP564I1171 , DKFZP586J0619, DUSP10, DUSP10, DYRK3, EEF2K, EFEMP1, EMP1, EVC, EVI2A, EXT2, FAM62A, FER1L3, FLJ14466, FLNA, FN1, FN1, GANAB, GDF15, GEM, GNPDA1, GPAA1, GPC6, GPNMB, GPNMB, GUSB, H2AFV, H2AFV, HDAC9, HDLBP, HECW2, HMGA2, HMOX1, HSDL2, HSPBAP1, HSPC196, HYOU1, IDS, IGFBP3, IKBKAP, INSIG1, IPO4, IRS2, JAG1, KDELR3, KIAA0251, KIAA0586, KIAA1211, KIAA1462, KIAA1706, KIAA1754, KRT18, KRT7, KRTAP4-7, LAMP2, LEPR, LEPREL1, LHFPL2, LIF, LOC286044, LOC339229, LOC90693, LRRC8E, MAFG, MAGED2, MCTP1, MGC16291, MGC19764, MGC5618, MRPS30, MRPS31, MTERFD3, MYH9, NAGA, NAV2, NCSTN, NEK9, NEU1, NFKBIZ, NMT2, NPC2, NSUN5C, NTNG1, NUP188, OACT2, OS9, P4HA1 , P8, PALM2-AKAP2, PALM2-AKAP2, PARVA, PBX2, PDE4DIP, PDIA4, PDIA6, PEG10, PHF19, PIK4CA, PLEKHM1, PLOD1, PLOD2, PPP1R15A, PPP1R15A, PRKDC, PRSS23, PRSS23, PSEN2, PSMD1, PTPRF, PTPRJ, RAB32, RAB9A, RG9MTD1, RGS4, RHOQ, RND3, RNF25, RNPEP /// UBE2V1 /// Kua /// Kua-UEV, RNU17D, ROBO4, RRAGC, RRS1, SEC31L1, SERPINB2, SERPINB7, SESN2, SGEF, SGSH, SKIV2L, SLC25A21, SLC35A3, SLC3A2, SMARCA1, SNAPC1, SNF1LK, SPOCD1, SPTAN1, SQSTM1, ST3GAL6, STC2, STX3A, TFPI2, TFPI2, TGFBI, TGM2, THRAP1, TLN1, TMEM60, TNFAIP3, TRIB3, TRIO, TSC2, UAP1L1, UBAP2L, UPP1, URB, USP11, USP5, VDR, WDR4, YTHDF2, ZCCHC9, ZDHHC20, ZFHX1B, ZNF185, ZNF278, ZNF690, ZNF697
Data obtained from our Microarray experiments disclose that Xanifolia Y inhibiting gene expression of the following genes (represented by gene symbol):
AKR1C3, SLC35A3, NEK9, CAV 1 , USP11, KRT7, TGM2, NCSTN, COG6, WDR4, GPAA1, GUSB, UBAP2L, NMT2, C9orf10, KIAA0251, BTD, EMP1, KRT18, OS9, CPSF1, PSMD1, RNPEP///UBE2V1 /// Kua /// Kua-UEV, NAGA, PARVA, HYOU1, ARMC4, APEH, BTD, FER1L3, CFH///CFHL1, COL5A2, EFEMP1, COL18A1, HSDL2, NUP188, IDS, PLOD1, CPM, SPTAN1, LAMP2, ARNTL2, PLOD2, KDELR3, KIAA0586, SMARCA1, PRSS23, PTPRJ, LEPREL1, H2AFV, CAD, URB, CPNE3, DKFZP586J0619, SERPINB7, CNAP1, EEF2K, IKBKAP, SLC25A21, KDELR3, PDIA6, CAV2, COL4A2, MAGED2, PHF19, OACT2, JAG1, FAM62A, KDELR3, PIK4CA, USP5, ZDHHC20, H2AFV, PTPRF, PEG10, P4HA1, MAGED2, PRSS23, PTPRF, CHRDL1, C6orf65, APLP2, EXT2, COPG, SKIV2L, PDIA4, MYH9, SEC31L1, GANAB, COL5A1 , ZNF185, DDB1, HDLBP, AP2B1, TSC2, IPO4, FLNA, TLN1, PRKDC, COL6A3, NTNG1, LEPR, RGS4, FN1, GPC6, LEPR, RGS4JGFBP3
Data obtained from our Microarray experiments disclose that Xanifolia Y stimulates gene expression of the following genes (represented by gene symbol):
P8, KRTAP4-7, DUSP10, CLDN14, ANGPT2, DUSP10, GDF15, GPNMB, HDAC9, HECW2, C14orf78, UPP1, PPP1R15A, PLEKHM1, STX3A, ANGPT2, SQSTM1, RHOQ, STC2, PPP1R15A, LOC286044, ATF3, HMOX1, CXCL2, CD33L3, LRRC8E, SESN2, LIF, TFPI2, KIAA1706, RRAGC, DDIT3, DYRK3, CTNS, GPNMB, CEBPB, CCL20, AMIGO2, KIAA1462, HSPBAP1, EVC, CLN8, ABL2, SGEF, MCTP1, IRS2, C14orf109, TNFAIP3, RND3, ZFHX1 B, LHFPL2, SNF1LK, SLC3A2, NAV2, SPOCD1, TFPI2, EVI2A, ST3GAL6, CSTB, ROBO4, GNPDA1, GEM, IRS2, HMGA2, PALM2- AKAP2, BRI3, KIAA1754, VDR, NEU1, INSIG1, C17orf32, ABL2, PALM2-AKAP2, MTERFD3, ZNF697, NFKBIZ, BCL6, THRAP1, MGC5618, ADAMTS1, MAFG, NPC2, CITED2, TRIB3, ZCCHC9, RNU17D, CITED2, RRS1 , NSUN5C, PBX2, RG9MTD1, SGSH, INSIG1, MGC16291, RAB9A, ARMCX6, SERPINB2, ATP6V1B2, DKFZP564I1171, ATP6V0E, HSPC196, MRPS30, ARMCX1, LOC339229, ANKRD11, C9orf103, PSEN2, ADAMTS1, SNAPC1, RNF25, ZNF278, TGFBI, UAP1L1, PDE4DIP, MGC19764, TMEM60, CEP4, KIAA1211, DDX20, CSRP2BP, ZNF690, TRIO, CLTA, ATP6V0E, RAB32, MRPS31, LOC90693, ATP6V1C1, CBLL1 , YTHDF2, FLJ14466, ARL10C, BCL2A1
Data obtained from our Microarray experiments disclose that Xanifolia YM(ACH-Y) stimulate gene expression of the following genes (represented by gene symbol): YM stimulate the gene: Gene Symbol
CXCL2, CSF2, IL6, NFKBIZ, CXCL3, EGR1, CLDN14, ATF3, IL1A, CSF3, PTGS2, NFKBIZ, KRTAP4-9, TNFAIP3, ABL2, KRTAP4-7, MMP3, DUSP10, FOS, DUSP1, PPP1R15A, DDIT3, DUSP10, HECW2, DSCR1, SESN2, TSLP, SLC7A11, HES1, SESN2, CXCL1, TAGLN3, HBEGF, IER3, RAB2, P8, IL8, GADD45A, LOC441461, GEM, LIF, KLF6, MGC4504, CCL20, PRB1 /// PRB2, THRA /// NR1D1, ERN1, NECAP2, DKK3, GNAO1, NFKBIA, CREB5, CLDN1, CDKN1A, PRDM1, SERPINB2, CD274, FST, LOC401317, BIRC3, PMAIP1, BMP2, IL24, BMP2, WHDC1L2, SGK, RND3, UPP1, C14orf34, ARID1B, KLF6, KLHL24, MAFF, IL12A, SAT, NSD1, JUNB, DUSP1, SLC3A2, ASNS, MAFF, PSAT1, CLTA, ZC3H12C, CLDN1, IL1B, ROBO4, RARA, BCL6, SLC7A11 , PDE4DIP, ZNF697, ATP2A2, ZFP36, REST, HDAC9, STAC2, L0C153222, ZC3H12A, SLC25A37, IL17C, TRIB3, CEBPB, PTX3, TRIB3, KIAA1718, ZNF385, TTMP, MGC5618, CSF2, DUSP5, SLC25A25, ADAMTS1, TFRC, SEC15L2, SCNN1A, HES1, LOC153222, C20orf72, ETS1, GDF15, KIAA1754, PACRG, PRSS22, TFRC, AQP2, STX3A, REM1, LIMK2, STC2, ISG20L1, BCL2A1 , HES4, TJP3, SLC7A5, CEBPG, LILRB2 /// LILRB3, LOC389429, LLGL1 , KLF6, CSNK1 E, DUSP1, ICAM1, PYY2, TPCN1, MFSD2, KIAA0690, SOD2, E2F7, GRIK5, PADM, HIP1R, IRAK2, EIF1, CEBPG, G0S2, KIAA0690, NEDD4L, MXD1, TXNDC5, PERLD1, HIF1A, RDH13, CTH, ETS2, FAH, CLK4, RIMS4, FLJ12355, LDLR, WARS, IFRD1, LRCH4, SLC1A4, FST, PPIF, ARHGEF2, PMAIP1, PBEF1, C14orf159, ATF4, FLJ90119, TGFBR3, DDX21, MUC8, TNFSF7, SPTLC2, KBTBD8, CIAS1, KIAA0476, NFKB1, DES /// FAM48A, ICAM1, INHBA, MARS, C1orf113, GRK6, TCF2, SLC7A1, TNFRSF10B, IER2, TFRC, SOS2, CARS, HAB1 /// LOC442496, KLHL24, SQSTM1, GIT1, IRS2, SARS, TRIM25, JUN, YRDC, ETS1, CTH, ABL2, CD274, FZD8, EREG, PFDN2, TMEM63B, DYRK3, GARS, RASSF8, CAMTA2, HOXC8, AARS, TBX3, RRAGC, BCAP29, TNFRSF10B, MCL1, PALM2-AKAP2, TMEM22, ZNF548, PCBP2, SLC1A4, FSTL3, TA-PP2C, PIM3, MAFG, NOL1, PEA15, ETS2, IQCG, BEX2, C12orf39, LDLR, VEGF, CCNL1, LOC440069, SNAPC1 , SLC27A5, MARS, ICAM1, AXUD1, KCNIP2, LOC146177 /// FLJ40941, GOT1, C11orf17 /// NUAK2, TTBK1, SNCA, NFRKB, GRB2, ADAMTS1, PALM2-AKAP2, CCNL1, JUN, SPRY4, SLC38A1 , E2F7, MGC27165, IDS, SLC7A1, FLJ11021, KCNK5, GYPC, MYC, CACNG6, PHLDA1, INSIG1, FLJ13448, LOC202051, GALNT10, TFEB, ABL2, MTHFD2, SERPINB8,TNPO2, MCF2L, SERTAD1, SPRY2, PHLDA1, SLC7A11, CXCL5, COMT, GTPBP2, RAB4B, ATXN7, OTUD5, VEGF, ADAMTS6, YARS, NAV3, SAMD4, C17orf32, SHMT2, CHIC2, ORAOV1, CREM, ALS2CL, AMIGO2, MGC19764, SFPQ, MGC11257, PLEKHM1, MMP28, SLC4A5, DNAJB9, ERRFI1, FZD8, MGC4399, GUCA1B, ATP13A3, KIAA0020, XBP1, ATP6V1B2, IGLV3-25, CHD4, GRB10, FLJ40089, CLDN19, NPC1, RIPK2, PLEKHF2, LOC283314, RSPO3, GSK3A, ANKRD11, WARS, GNPDA1, IGKC /// IGKV1-5, RCL1 , C9orf91, TIMM10, SLC1A4, ODC1, SLC38A1, EIF4EBP1, ZNF267, YRDC, CHD4, PER1, NFKBIE, INSIG1, FLJ11259, NCOA3, TNPO2, BAZ2A, SLC1A5, KRTAP2-1, PDE4DIP, EPB41L3, CITED2, JTV1 , TA-PP2C, METRNL, BCL10, C1orf19, PTP4A1 , PDE4DIP, CPSF1 , TIMM10, IGSF21, TM4SF1 , CDK6, BANP, PHLDA1, PAPPA2, MGC17337, TNFRSF19L, ADAMTS10, CBFB, HERPUD1, RTTN, FLJ13855, SYNPO, NAV2, FLJ34208, DDX10, C6orf66, LOC56902, CHMP1B, HIPK2, H3F3B, NAV3, ELL2, NSUN5C, PLAUR, UCK2, PBEF1, TP53BP2, TEX10, B3GNT5, USP36, INHBA, SPIRE1, CSTB, NSUN5C, TFPI2, Our experiment disclosed that Xanifolia Y and ACH-Y inhibited genes expression of the following genes: FN1, ITGAV, LAMA4, LAMB2, LAMC1, LAMB1, LAMB1, LAMA4, LAMA5, LAMC1, LAMA2, LAMB1, LAMA3, SCAMP1, TICAM2, SCAMP1, TICAM2, SCAMP1, SCAMP1, CAMK2B, DL1 , ICAMS1CEECAMIJCAMS1SCAMPI, CAMK1G, CAMSAP1, MCAM, CAMTA1, CKN1, ALCAM, DCAMKL2, CEACAM3, CAMK2D, CAMK2B, SCAMP5, CAMK4, NCAM1, CAMK2G, MYH9, MYH10, MY01 D, MYO5A, MYLK, MYO6, MYO5A, MYO1C, MYLK, MY06, MYLC2PL, MYO10, MY06, TPM3, MYO1C, BECN1, MYO1 E, TPM3, M-RIP, MYO1 B, MYO10, MYO5A, M-RIP, MYO10, MYL6, MYOHD1, BECN1, TPM4, MYLK, MYH10, MYOHD1, LOC221875, LOC402643, MYO15B, LOC129285, MYH11 , MY01 B, MYO1 C, MYO9B, CDH13, CTNNAL1 , CDH13, CDH12, CTNNB1, CDH5, CTNND1, CDH2, CTNNA1, CDH2, PCDHB16, CTNNA1, CELSR2, PCDHB6, PCDHB7, CTNND2, PCDHGC3, PCDHGB4, PCDHGA8, PCDHGA12, PCDHGC5 , PCDHGC4 , PCDHGB7, PCDHGB6, PCDHGB5, PCDHGB3, PCDHGB2, PCDHGB1, PCDHGA11, PCDHGA10 , PCDHGA9, PCDHGA7, PCDHGA6, PCDHGA5, PCDHGA4 , PCDHGA3 , PCDHGA2, PCDHGA1, CTNND1, CDH23, PCDHB12, PCDHB10, PCDH18, CDH20, PCDH9, PCDHGA12 , PCDHGA11, PCDHGA10, PCDHGA6, PCDHGA5, PCDHGA3, PCDH7, CDH18, CDH6, CCBE1, COL10A1, COL12A1, COL13A1, COL18A1, COL1A1, COL21A1, COL4A1 , COL4A2, COL4A5, COL4A6, COL5A1 , COL5A2, COL6A1, COL6A2, COL6A3, COL9A1, MMP9, P4HA1, P4HA2, P4HB, PCOLCE, PCOLCE2, PCOTH, PL0D1, PL0D2, PL0D3, CIB1, ILK, ITGA2, ITGA3, ITGA4, ITGA6, ITGAV, ITGB1, ITGB1BP1, ITGB2, ITGB5, ITGBL1 , TNC, EMILIN1, ICAM1, HSPG2, HPSE, HS2ST1.SDC2,
Determination of gene expression by Real-time PCR method (Brilliant QPCR, Agilent Technologies): The real-time polymerase chain reactions further confirm the results obtained from micro array analysis. The Real-time PCR results (shown below) confirmed that Compound Y3 and YM increase the expression of the genes: ANGPT2, DDIT3, LIFandNFKBIZ Table 19:
ANG PT2 DDIT3 LIF NFKB1Z
% of control
Control 100 100 100 100
Y3 300 213 163 142.9
YM 216 660 447 784.8
YM = Y3 without
D = DMSO control Y = Y3 sugar
Table 20: The experimental results confirmed that compound Y3 inhibit the gene expression of adhesion proteins: ITGAV, TNC, COL1 A1 , FN 1 and LAMA4
ITGAV TNC COL1A1 FN1 LAMA4
% of control
Control 100 100 100 100 100
Y3 63.8 41 .1 49.9 44.1 53.6
With the same method, we also confirmed that the compound YM inhibits the expression of adhesion proteins COL1 A1 , FN1 and LAMA4
Table 21 :
COL1A1 FN1 LAMA4
% of control
Control 100 100 100
YM 42 72.8 81
ANGPT2 =angeopoietin 2, LIF = Leukemia inhibitory factor
ITGAV = integrin alpha V, (cholinergic differentiation factor),
TNC = Tenascin C, NFKB1 Z = nuclear factor of kappa light
COL1 A1 = collagen type 1 alpha 1 , polypeptide gene enhancer in B-cells
FN1 = fibronectin 1 , inhibitor, zeta
LAMA4 = laminin alpha 4,
DDIT3 = DNA-damage-inducible transcript
3,
Data obtained from our Microarray experiments disclose that Xanifolia Y modulates gene expression.This invention provides compositions and methods for modulating the gene expression in cancer cells, wherein the modulating comprises of positive and negative regulation, wherein genes being modulatated are adhesion proteins; wherein modulation includes expression, production and secretion of adhesion proteins, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin , vitronectin, collagen, laminin, cadherin, heparin, Glypican, tenascin, CD 54, CAM. This invention provides compositions and methods for modulating angiopoietins, wherein comprises positive regulating the angiopoietin 2, wherein comprises negative regulating angiopoietin 1 . The composition and method of this invention comprises a triterpene wherein acylation group at carbon position 21 and/or 22 of the triterpene is necessary for the function and are selected from angeloyl, acetyl, alkanoyl, and alkenoyl and acyl group. The sugar moiety (ies) at position 5 of the triterpene is important for enchancing activity of these compounds.
Leptin is a promoter in breast cancer cells. The Xanifolia Y and YM inhibits the expression of leptin.
The thrombospondins are a family of multifunctional proteins. The family includes thrombospondins 1 -5. Thrombospondin-1 TSP1 is a multifunctional, matricellular glycoprotein, containing interacting domains for a large variety of adhesion, migration and proliferation, angiogenesis and tumor cell metastasis.
The insulin-like growth factor binding protein (IGFBP) is a multifunctional protein that relate to metastasis, tumor growth and angiogenesis. Compounds in this application modulate gene expression of RAB3B, inhibiting metastasis, tumor growth and angiogenesis.
The activation of cell migration and cancer cells metastasis requires the sensitive of potassium channel. The results of the micro array experiment showed that compound Y and YM inhibit gene expression of family protein relate to potassium channel in ES2 cells.
RAB3B is member RAS oncogene family protein. Compounds in this application modulate gene expression of RAB3B, inhibiting metastasis, tumor growth and angiogenesis.
In mild conditions, the saponin can be partially hydrolyzed into a mixture of products which are separated by HPLC. Specific partial hydrolysis of saponins can also be achieved with enzymes. The glycosidases catalyze the hydrolysis of the glycosidic linkage. Galactosidase is an enzyme which catalyzes the hydrolysis of galactosides. The glucosidase is an enzyme which breaks glucose from saponin. Other enzyme examples are xylanases, lactase, amylase, chitinase, sucrase, maltase, and neuraminidase.
The sugar moiety of the triterpenoid saponin (example Xanifolia Y) can be removed by acid hydrolysis. The synthetic compound of ACH-Y is obtained. The ACH-Y is a triterpene with acyl groups but no sugar moiety. The acyl group of the saponin (example Xanifolia Y) can be removed by alkaline hydrolysis. A synthetic compound of AKOH-Y can be obtained. The AKOH-Y is a pentacyclic triterpene with sugar moieties. A pentacyclic triterpene can be obtained by acid and alkaline hydroysis of saponin from natural source. A pentacyclic triterpene also can be obtained by other methods (Reference: Surendra et al., Rapid and Enantioselective Synthetic Approches to Germanicol and Other Pentacyclic Triterpenes). The pentacyclic triterpene with sugar moieties can also be obtained with synthesis (Reference: PIe et al., Synthesis of L- arabinopyranose containing hederagenin saponins). Acylation is the process of adding an acyl group to a compound. Friedel-Crafts reaction is an example of the process. An active compound can be obtained by acylating a pentacyclic triterpene. In an embodiment, acylating G21 and C22 of a pentacyclic triterpene gives an anticancer compound . In an embodiment, sugar moiety(s) at C3 can increase the activities of pentacyclic triterpene, wherein the triterpene has acyl group(s), wherein the acyl group(s) may be at C21 , 22, or 28. In an embodiment, a sugar moiety is at C21 , 22, or 28, wherein the sugar moiety is substituted with 2 acyl groups. Reference in PCT/US/US05/319000, WO20061029221 , filed September 7, 2005, US1 1 /289142 filed November 28, 2005)
Determination of cell growth of Leishmania parasites by MTT assay shows that Y10 is cytotoxic to Leishmania Major (promastigotes) with IC50 of 15ug/ml. IC 50 of Y is 15ug/ml, YO is 25ug/ml, Y1 is 23ug/ml, Y5 is 16ug/ml, Y7 is 18ug/ml, ACH-Y Is 30ug/ml, Mb5 is 15ug/ml, ACH-Mb5 is 18ug/ml, Z12 is 23ug/ml, and Ba1 is 15ug/ml
EXPERIMENTAL DETAILS
Experiment details of herb extraction, analysis of extract components by HPLC, determination of the cell-growth activity effected by Xanifolia Y with cells derived from different human organs using MTT Assay, purification of the bioactive components from plant extract, fractionation of plant extracts with FPLC, isolation of component Ys with preparative HPLC, determination of the chemical structure, cell experiments and animal studying are disclosed in PCT/US05/31900, U.S. Serial No. 1 1 /289142, U.S. Serial 10/906303, U.S. Serial No. 1 1/131551 and U.S. Serial Nos.1 1/683198, filed on March 7, 2007, PCT/US2007/077273, filed August 30, 2007, U.S. Serial No. 60/890380, filed on February 16, 2007, U.S. Nos. 60/947,705, filed on July 3, 2007, PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008, the contents of which are incorporated herein by reference. Experiments 1 -23 of PCT/US2008/002086, 1 188-ALA-PCT, filed February 15, 2008 are incorporated herein by reference.
Experiment 1 : Removal of the sugar moiety from saponin by acid hydrolysis
15 mg saponin was dissolved in 1 ml of Methanol. 1 ml of 2N HCI was then added. The mixture was refluxed in 8OC water bath for 5 hours. The solution was then neutralized by adding 2 ml of 1 N NaOH (to final pH 4-6). The aglycone was then extracted with ethylacetate 3 ml x 2. The extracts were collected and pooled. Further isolation of aglycone (sugar-removed saponin) was achieved by HPLC with isocratic elution of 80-100% acetonitrile. Experiment 2: Removal of the acyl group by alkaline hydrolysis Methods: 20 mg of saponin was dissolved in 0.5 ml of 1 M NaOH. The solution was incubated in 8OC water bath for 4 hours. It was cooled to room temperature before neutralized with 0.5 ml 1 N HCI (adjust pH to about 3). The mixture was extracted with 2 ml 1 -butanol 3 times. The butanol fractions were collected and lyophilized. The hydrolyzed saponin with further purified with HPLC in a C-18 column eluted with 25% acetonitrile.
Experiment 3: Analysis of genesis of blood vessel in xenograft tumor treated with compound Y
Methods: Athymic Nu/Nu mice (5-6 weeks old) are divided into two groups (1 and 2) with 5 animals in each group. On day 0, all mice were transplanted intra-peritoneally with one million ES2 (human ovarian cancer) cells. Animals were randomly divided into two groups: Group 1 : Control group. Animals did not receive drug-treatment.
Group 2: Drug-treatment group. On days 10-15 and 18-22, animals received daily drug administration of Xanifolia-Y, by i.p. route at dosage of 2.5 mg/kg.
Results: Animals showed high tumor burden after 18 days. Animals with high tumor burden were euthanized and solid tumors were taken out from these mice (between 18-
27 days). Tumor tissues fixed with formalin at room temperature. The fixed tissues were sectioned and stained with Haematoxylin and eosin (H&E). The red blood cells inside the micro blood vessels were identified under a microscope. Figure 1 shows that more blood vessels were observed in the control Groupi than those in the drug-treated Group 2
Experiment 4: Determination of cell growth of Leishmania parasites by MTT assay.
Leishmania parasites (Leishmania major: MRHO/SU/59/P/LV39) were grown in culture medium in a T75 flask at room temperature. Promastigotes of Leishmania major (approximately 40 million per ml) were used in the experiment. 1 .2 ml cell culture was transferred to a well of the 24-wells plate. Saponin Y10 (0.2 ml in medium) with different concentrations (final 6.25 -200 ug/ml) was added to culture and cells were grown for 1 -5 days at room temperature. At the end of drug-treatment, 150 ul of MTT (5 mg/ml in PBS) was added to each well and incubated for 4 hours. Formazan formed in cells was dissolved with DMSO and the OD at 490nm was determined by an ELISA reader. Results: this experiment shows that Y10 is cytotoxic to Leishmania Major (promastigotes) with IC50 approximately equal to 15ug/ml. Experiments are repeated with Y, ACH-Y, AKOH-Y, Mb5, ACH-Mb5, AKOH-Mb5 and Ba1 .

Claims

What is claimed is:
1 . A method for modulating adhesion protein of cells, inhibiting angiogenesis in tumor, modulating the gene expression, modulating the angiopoietin, enhancing an immune response, providing adjuvant activities, providing vaccine activities, or inhibiting the metastasis of cancer cell in a subject, comprising contacting said subject with an effective amount of an isolated, purified or synthesized compound, or its salt, or ester thereof, selected from the formula:
, also named (1 E), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O- alkyl, 0-dιbenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, 0-heterocylιc, 0-heteroraryl, and derivatives thereof; R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O- alkyl, 0-dιbenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, 0-heterocylιc, 0-heteroraryl, and derivatives thereof; R4 represents CH3, CHO, CH2R6 or COR6, wherein R6 is selected from hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, 0-dιbenzoyl, O-benzoyl, O- alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, 0-heterocylιc,
O-heteroraryl, and derivatives thereof; R3 is H or OH; R8 is H or OH, preferably OH;
R16 is H ,or R4 and R16 may form an oxygen bridge with divalent radical formula of -CH2-O-, CH(OH)-O- or C(=0)-0-, wherein the -O- may be replaced with - N- ; wherein when the C12-13 of ring 3 of the tπterpene has a double bond then
R16 is absent,
R5 is a hydrogen, heterocyclic or sugar moiety(ιes), wherein the sugar moιety(ιes) is/are selected from a group consisting of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, πbose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, and derivatives or combination thereof; wherein R9, R10, R1 1 , R12, R13, R14, R15 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO-alkyl, COO-aryl, COO- heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocyclic, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; wherein at least two of R1 , R2 and R6 are compriseing a group selected from O-angeloyl, O-tigloyl, O- senecioyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; or at least one of R1 , R2, and R4 is a sugar moiety substituted with at least two groups selected from a group consisting of angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, and a derivative thereof; or wherein R4 is CH2R6, wherein R1 and R2 independently consists an O- angeloyl group, or at least two of R1 , R2 and R6 are O-angeloyl or at least one of R1 , R2 or R6 is a sugar moiety with two O-angeloyls; or wherein R5 is/are the sugar moiety(ies) selected from the following sugars and alduronis acids: glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, glucuronic acid, galacturonic acid; or their derivatives thereof, or the combination thereof; wherein the sugar preferably comprises glucuronic acid, arabinose and galactose; or wherein R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof; or wherein R5 is 3-β- 0-{[(α-L-rhamnopyranosyl-(1 →2)]-α-L-rhamnopyranosyl--(1 →2)-β-D- galactopyranosyl--(1 →3)]-[β-D- galactopyranosyl~(1 →2)]- β-D- glucuronopyranosyl}
2. The method of claim 1 , wherein at least one of R1 and R2 of the compound is selected from O-acetyl, O-angeloyl, O-tigloyl, O-senecioyl, O-dibenzoyl, and O- benzoyl, or at least one of R1 and R2 is a sugar moiety substituted with two groups selecting from acetyl, angeloyl, tigloyl, senecioyl, dibenzoyl, benzoyl; R5 is a hydrogen or sugar moiety(ies), wherein the sugar moiety(ies) is/are selected from a group of glucose, galactose, arabinose and derivatives thereof, wherein the derivatives are acid, ester and salt.
3. The method of claim 1 , wherein the modulating adhesion of cancer cell comprises inhibiting the secretion or expression of adhesion protein, wherein the adhesion proteins comprise fibronectin, integrins family, Myosin, vitronectin, collagen, laminin, polyglycans, cadherin, heparin, tenascin, CD 54 and CAM; wherein the inhibiting protein expressed from genes, including ITGAV, TNC, COL1 A1 , FN1 , LAMA4, family protein relate to potassium channel, RAB3B, thrombospondin, insulin-like growth factor, G-protein and Glypican.
4. The method of claim 1 , wherein the modulating adhesion protein comprises reducing the fibronectin for inhibiting the metastasis or growth of cancer cells, wherein the cancer is selected from breast, leukocyte, liver, ovarian, bladder, prostate, skin, bone, brain, leukemia, lung, colon, CNS, melanoma, renal, cervix, esophagus, testis, spleen, kidney, lymph, pancreas, stomach and thyroid cancer.
5. The method of claim 1 , wherein the inhibiting angiogenesis comprises modulating angiopoietin comprising angiopoietin 1 , angiopoietin 2, angiopoietin 3, angiopoietin 4, angiopoietin 5, angiopoietin 6 and angiopoietin 7; or wherein the angiopoietin is comprising angiopoietin-like 1 , angiopoietin-like 2, angiopoietin - like 3, angiopoietin-like 4, angiopoietin-like 5, angiopoietin-like 6 and angiopoietin-like 7; or wherein the modulating comprises positive and negative regulating; wherein modulating angiopoietin comprises stimulating the angiopoietin 2 in order to inhibit angiogenesis; or wherein modulating angiopoietin comprises inhibiting the angiopoietin 1 in order to inhibit angiogenesis; or wherein modulating angiopoietin comprises inhibiting the angiopoietin-like 1 ; or wherein modulating angiopoietin comprises inhibiting the angiopoietin-like 4.
6. The method of claim 1 , wherein the modulating gene expression includes increasing the expression of the protein phosphatase 1 , dual specificity phosphatase 10; or increasing expression of the genes of ANGPT2, DDIT3, LIF and NFKB1 Z.
7. The use of composition of claim 1 , wherein the compound is selected from the following: a) An isolated, purified or synthesized compound is having structure Xanifolia(Y), or chemical name: 3-0-[β-D- galactopyranosyl (1 →2)]-α-l_-arabinofuranosy (1 →3)-β-D-glucuronopyranosyl- 21 ,22-0-diangeloyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
b) An isolated, purified or synthesized compound is having structure Xanifolia (Y1 ).
chemical name: 3-0-[β-D-galactopyranosyl (1 →2)]-α-l_-arabinofuranosyl (1 →3)-β-D-glucuronopyranosyl-21 -O-(3,4- diangeloyl)-α-L-rhamnophyranosyl-22-0-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene;
c) An isolated, purified or synthesized compound is having structure Xanifolia (Y2),
or chemical name: 3-0-[β-D-glucopyranosyl- (1 →2)]-α-L-arabinofuranosy (1 →3)-β-D-glucuronopyranosyl-21 ,22-O-diangeloyl- 3β, 15α, 16α, 21 β, 22α, 24β, 28-heptahydroxyolean-12-ene;
d) An isolated, purified or synthesized compound is having structure Xanifolia (Y8), or chemical name: 3-0-[/?-glucopyranosyl (1 →2)]-α-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 , 22-0-diangeloyl-3/?, 16a, 2λβ, 22a, 2Λβ, 28-hexahydroxyolean-12-ene;
e) An isolated, purified or synthesized compound is having structure Xanifolia (Y9).
or chemical name: 3-0-[β- galactopyranosyl (1 →2)]-α--arabinofuranosyl (1 →3)-/5-glucuronopyranosyl-21 -O- (3,4-diangeloyl)-α-rhamnopyranosyl-28-0-acetyl-3^, 16a; 21^, 22a, 28- pentahydroxyolean-12-ene; and
f) An isolated, purified or synthesized compound is having structure Xanifolia (Y10),
, or chemical name:
3-0-[/?-galactopyranosyl (1 →2)]-α-arabinofuranosyl (1 →3)-/5-glucuronopyranosyl- 21 , 22-0-diangeloyl-3# 16«, 2Λβ, 22a, 28-pentahydroxyolean-12-ene.
g) An isolated, purified or synthesized compound is having structure Xanifolia (YO), or chemical name: 3-0-[β-D- galactopyranosyl(1 →2)]-α-l_-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl-21 - O-angeloyl, 22-0-(2-methylpropaπoyl)-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene,
h) An isolated, purified or synthesized compound is having structure Xanifolia (X),
, or chemical name: 3-0-{[β-D- galactopyranosyl (1 →2)]-[αr-l_-arabinofuranosyl (1 →3)]- /2-D-glucuronopyranoside butyl ester}-21 -O-acetyl-22-O- angeloyl- 3y5,16α,21y5,22αζ28-pentahydroxyolean- 12-ene.
i) An isolated, purified or synthesized compound is having structure (Y7),
or chemical name: 3-0-[/3-D- galactopyranosyl-(1 →2)]-σ-L-arabinofuranosyl-(1 →3)-/3-D--glucuronopyranosyl- 21 -0-angeloyl-28-0-2-methylbutanoyl-3)8, 15 σ, 16σ, 21 /3, 22σ, 28- hexahydroxyolean-12-ene
j) An isolated, purified or synthesized compound is having structure (ACH-Y): k) An isolated, purified or synthesized compound is having structure:
or chemical name:
3-O-[/?-glucopyranosyl (1 →2)]- /?-arabinofuranosyl {\ →4)-β- glucuronopyranosyl -21 -O-angeloyl-22-O-acteyl-3# 16a, 2λβ, 22a, 24β, 28-hexahydroxyolean-12- ene;
(I) An isolated, purified or synthesized compound is having structure:
(m) An isolated, purified or synthesized compound is having structure:
wherein R1 , R2, are O(C=O)C(CH3)=CH(CH3), R3 is OH, R4 is OH (n) An isolated, purified or synthesized compound is having structure:
(o) An isolated, purified or synthesized compound is having structure:
,wherein R1 is O(C=O)C(CH3)=CH(CH3), R2 is O(C=O)CH=CH-C6H5, R3, R4 are OH, R6, R7, R8, R9, R10, R1 1 , R12 are CH3, R13 is COOCH3, also named Mb12 (p) An isolated, purified or synthesized compound is having structure:
3-0-[β-D-galactopyranosyl (1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-O-angeloyl-3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba 1 ;
3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0- tigloyl -3β, 16α, 21 β, 22α, 28- pentahydroxyolean-12-ene;
8. A method for treating parasites in a subject, comprising contacting said subject with an effective amount of an isolated, purified or synthesized compound, or its salt, or ester thereof, selected from the formula: , also named (1 E), wherein R1 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O- alkyl, 0-dιbenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof;
R2 is selected from hydrogen, hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O- alkyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; R4 represents CH3, CHO, CH2R6 or COR6, wherein R6 is selected from hydroxyl, O-angeloyl, O-tigloyl, O-senecioyl, O-alkyl, O-dibenzoyl, O-benzoyl, O- alkanoyl, O-alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; R3 is H or OH; R8 is H or OH, preferably OH;
R16 is H ,or R4 and R16 may form an oxygen bridge with divalent radical formula of -CH2-O-, CH(OH)-O- or C(=0)-0-, wherein the -O- may be replaced with - N- ; wherein when the C12-13 of ring 3 of the tπterpene has a double bond then R16 is absent; wherein the sugar moieties can not be 5, if the oxygen bridge is formed between R4 and R16, R5 is a hydrogen, heterocyclic or sugar moιety(ies), wherein the sugar moiety(ιes) is/are selected from a group consisting of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, alduronic acid, glucuronic acid, galacturonic acid, and derivatives or combination thereof; wherein R9, R10, R1 1 , R12, R13, R14, R15 are independently attached a group selecting from CH3, CH2OH, CHO, COOH, COO- alkyl, COO-aryl, COO-heterocyclic, COO-heteroaryl, CH2Oaryl, CH2O- heterocychc, CH2O- heteroaryl, alkyls group, hydroxyl, acetyl group, particularly CH3; wherein at least two of R1 , R2 and R6 are compriseing a group selected from O-angeloyl, O-tigloyl, O-senecioyl, O-dibenzoyl, O-benzoyl, O-alkanoyl, O- alkenoyl, O-benzoyl alkyl substituted O-alkanoyl, O-aryl, O-acyl, O-heterocylic, O-heteroraryl, and derivatives thereof; or at least one of R1 , R2, and R4 is a sugar moiety substituted with at least two groups selected from a group consisting of angeloyl, acetyl, tigloyl, senecioyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, and a derivative thereof; or wherein R4 is CH2R6; wherein R1 and R2 independently consists an O-angeloyl group, or at least two of R1 , R2 and R6 are O-angeloyl or at least one of R1 , R2 or R6 is a sugar moiety with two O- angeloyls; or wherein R5 is/are the sugar moiety(ies) selected from the following sugars and alduronis acids: glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, glucuronic acid, galacturonic acid; or their derivatives thereof, or the combination thereof; wherein the sugar preferably comprises glucuronic acid, arabinose and galactose; or wherein R5 is/are sugar moiety(ies) selected from a group consisting of glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid, and a derivative or combination thereof; or wherein R5 is 3-β-O-{[(α-L-rhamnopyranosyl-(1 →2)]-α-L-rhamnopyranosyl-- (1 →2)-β-D- galactopyranosyl--(1 →3)]-[β-D- galactopyranosyl-(1 →2)]- β-D- glucuronopyranosyl}
9. The method of claim 8, wherein at least one of R1 and R2 is selected from O- acetyl, O-angeloyl, O-tigloyl, O-senecioyl, O-dibenzoyl, and O-benzoyl, O- alkanoyl substituted phenyl, O-alkenoyl substituted phenyl, or at least one of R1 and R2 is a sugar moiety substituted with two groups selecting from acetyl, angeloyl, tigloyl, senecioyl, dibenzoyl, benzoyl O-alkanoyl substituted phenyl, O- alkenoyl substituted phenyl; R5 is a hydrogen or sugar moiety(ies), wherein the sugar moiety(ies) is/are selected from a group consisting of glucose, galactose, arabinose and derivatives thereof, wherein the derivatives are acid, ester and salt.
10. The method of claim 8, wherein the antiparasitics comprise inhibiting leishmaniases, amoebiasis, trypanosomiasis, toxoplasmosis or malaria.
1 1 . The method of claim 8, wherein the compound is selected from the following: a) An isolated, purified or synthesized compound is having structure Xanifolia(Y), chemical name: 3-0-[β-D-galactopyranosyl
(1 →2)]-α-l_-arabinofuranosy (1 →3)-β-D-glucuronopyranosyl-21 ,22-O-diangeloyl- 3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene;
b) An isolated, purified or synthesized compound is having structure Xanifolia
(Y1 ).
or chemical name: 3-0-[β-D-galactopyranosyl
(1 →2)]-α-L-arabinofuranosyl (1 →3)-β-D-glucuronopyranosyl-21 -O-(3,4- diangeloyl)-α-L-rhamnophyranosyl-22-0-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene;
c) An isolated, purified or synthesized compound is having structure Xanifolia (Y2),
chemical name: 3-0-[β-D-glucopyranosyl- (1 →2)]-α-l_-arabinofuranosy (1 →3)-β-D-glucuronopyranosyl-21 ,22-O-diangeloyl-
3β, 15α, 16α, 21 β, 22α, 24β, 28-heptahydroxyolean-12-ene;
d) An isolated, purified or synthesized compound is having structure Xanifolia (Y8), or chemical name: 3-0-[/?-glucopyranosyl (1 →2)]-c^arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 , 22-0-diangeloyl-3/?, 16a, 2λ β, 22a, 2Aβ, 28-hexahydroxyolean-12-ene;
e) An isolated, purified or synthesized compound is having structure Xanifolia (Y9).
chemical name: 3-0-[/?-galactopyranosyl (1 →2)]-c^arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 -O-(3,4-diangeloyl)-or- rhamnopyranosyl-28-0-acetyl-3/?, 16a, 2λβ, 22a, 28-pentahydroxyolean-12-ene; and
f) An isolated, purified or synthesized compound is having structure Xanifolia (Y10),
, or chemical name:
3-0-[/?-galactopyranosyl (1 →2)]-α-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl- 21 , 22-0-diangeloyl-3yff, ^\ 6a, 2λ β, 22a, 28-pentahydroxyolean-12-ene.
g) An isolated, purified or synthesized compound is having structure Xanifolia (YO), or chemical name: 3-0-[β-D- galactopyranosyl(1 →2)]-α-l_-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl-21 - O-angeloyl, 22-0-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene,
h) An isolated, purified or synthesized compound is having structure Xanifolia (X),
or chemical name: 3-0-{[/?-D- galactopyranosyl (1 →2)]-[«-L-arabinofuranosyl (1 →3)]-/5-D-glucuronopyranoside butyl ester}-21 -0-acetyl-22-0-angeloyl- 3y?,16α,2i y?,22α,28-pentahydroxyolean- 12-ene.
i) An isolated, purified or synthesized compound is having structure (Y7),
or chemical name: 3-0-[/3-D- galactopyranosyl-(1 →2)]-σ-L-arabinofuranosyl-(1 →3)-/3-D--glucuronopyranosyl- 21 -0-angeloyl-28-0-2-methylbutanoyl-3/3, 15 σ, 16σ, 21 /3, 22σ, 28- hexahydroxyolean-12-ene
j) An isolated, purified or synthesized compound is having structure (ACH-Y):
k) An iso compound is having structure:
or chemical name:
3-0-[/k)lucopyranosyl (1 →2)]- /?-arabinofuranosyl (1 →4)-/?-glucuronopyranosyl- 21 -0-angeloyl-22-0-acteyl-3# 16a; 2Λβ, 22a, 2Λβ, 28-hexahydroxyolean-12- ene;
m)
wherein R1 , R2, are
O(C=O)C(CH3)=CH(CH3), R3 is OH, R4 is OH (n)
(o)
3-O-[β-D-galactopyraπosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0-angeloyl-3β, 15α, 21 β, 22α, 28- pentahydroxyolean-12-ene, named Ba 1 ; (P)
3-0-[β-D-galactopyranosyl(1 →2)]-β- D-xylopyranosyl (1 →3)-β-D- glucuronopyranosyl-21 -O-angeloyl, 22-0- tigloyl -3β, 16α, 21 β, 22α, 28- pentahydroxyolean-12-ene;
12. The method of claim 1 , wherein the expression of the following genes is inhibited (represented by gene symbol) :
AKR1 C3, SLC35A3, NEK9, CAV1 , USP1 1 , KRT7, TGM2, NCSTN, COG6, WDR4, GPAA1 , GUSB, UBAP2L, NMT2, C9orf10, KIAA0251 , BTD, EMP1 , KRT18, OS9, CPSF1 , PSMD1 , RNPEP///UBE2V1 /// Kua /// Kua-UEV, NAGA, PARVA, HYOU1 , ARMC4, APEH, BTD, FER1 L3, CFH /// CFHL1 , COL5A2, EFEMP1 , COL18A1 , HSDL2, NUP188, IDS, PLOD1 , CPM, SPTAN1 , LAMP2,
ARNTL2, PLOD2, KDELR3, KIAA0586, SMARCA1 , PRSS23, PTPRJ, LEPREL1 , H2AFV, CAD, URB, CPNE3, DKFZP586J0619, SERPINB7, CNAP1 , EEF2K, IKBKAP, SLC25A21 , KDELR3, PDIA6, CAV2, COL4A2, MAGED2, PHF19, OACT2, JAG1 , FAM62A, KDELR3, PIK4CA, USP5, ZDHHC20, H2AFV, PTPRF, PEG10, P4HA1 , MAGED2, PRSS23, PTPRF, CHRDL1 , C6orf65, APLP2, EXT2, COPG, SKIV2L, PDIA4, MYH9, SEC31 L1 , GANAB, COL5A1 , ZNF185, DDB1 , HDLBP, AP2B1 , TSC2, IPO4, FLNA, TLN1 , PRKDC, COL6A3, NTNG1 , LEPR,
RGS4, FN1 , GPC6, LEPR, RGS4JGFBP3; or wherein the expression of the following genes are stimulated (represented by gene symbol): P8, KRTAP4-7, DUSP10, CLDN14, ANGPT2, DUSP10, GDF15, GPNMB, HDAC9, HECW2, C14orf78, UPP1 , PPP1 R15A, PLEKHM1 , STX3A, ANGPT2, SQSTM1 , RHOQ, STC2, PPP1 R15A, LOC286044, ATF3, HMOX1 , CXCL2,
CD33L3, LRRC8E, SESN2, LIF, TFPI2, KIAA1706, RRAGC, DDIT3, DYRK3, CTNS, GPNMB, CEBPB, CCL20, AMIGO2, KIAA1462, HSPBAP1 , EVC, CLN8, ABL2, SGEF, MCTP1 , IRS2, C14orf109, TNFAIP3, RND3, ZFHX1 B, LHFPL2, SNF1 LK, SLC3A2, NAV2, SPOCD1 , TFPI2, EVI2A, ST3GAL6, CSTB, ROBO4, GNPDA1 , GEM, IRS2, HMGA2, PALM2-AKAP2, BRI3, KIAA1754, VDR, NEU1 ,
INSIG1 , C17orf32, ABL2, PALM2-AKAP2, MTERFD3, ZNF697, NFKBIZ, BCL6, THRAP1 , MGC5618, ADAMTS1 , MAFG, NPC2, CITED2, TRIB3, ZCCHC9, RNU17D, CITED2, RRS1 , NSUN5C, PBX2, RG9MTD1 , SGSH, INSIG1 , MGC16291 , RAB9A, ARMCX6, SERPINB2, ATP6V1 B2, DKFZP564I1 171 , ATP6V0E, HSPC196, MRPS30, ARMCX1 , LOC339229, ANKRD1 1 , C9orf 103,
PSEN2, ADAMTS1 , SNAPC1 , RNF25, ZNF278, TGFBI, UAP1 L1 , PDE4DIP, MGC19764, TMEM60, CEP4, KIAA121 1 , DDX20, CSRP2BP, ZNF690, TRIO, CLTA, ATP6V0E, RAB32, MRPS31 , LOC90693, ATP6V1 C1 , CBLL1 , YTHDF2, FLJ14466, ARL1 OC, BCL2A1 ; or wherein the expression of the following genes are stimulated (represented by gene symbol): CXCL2, CSF2, IL6, NFKBIZ,
CXCL3, EGR1 , CLDN14, ATF3, IL1 A, CSF3, PTGS2, NFKBIZ, KRTAP4-9, TNFAIP3, ABL2, KRTAP4-7, MMP3, DUSP10, FOS, DUSP1 , PPP1 R15A, DDIT3, DUSP10, HECW2, DSCR1 , SESN2, TSLP, SLC7A1 1 , HES1 , SESN2, CXCL1 , TAGLN3, HBEGF, IER3, RAB2, P8, IL8, GADD45A, LOC441461 , GEM, LIF, KLF6, MGC4504, CCL20, PRB1 /// PRB2, THRA /// NR1 D1 , ERN1 ,
NECAP2, DKK3, GNAO1 , NFKBIA, CREB5, CLDN1 , CDKN1 A, PRDM1 , SERPINB2, CD274, FST, LOC401317, BIRC3, PMAIP1 , BMP2, IL24, BMP2, WHDC1 L2, SGK, RND3, UPP1 , C14orf34, ARID1 B, KLF6, KLHL24, MAFF, IL12A, SAT, NSD1 , JUNB, DUSP1 , SLC3A2, ASNS, MAFF, PSAT1 , CLTA, ZC3H12C, CLDN1 , IL1 B, ROBO4, RARA, BCL6, SLC7A1 1 , PDE4DIP, ZNF697,
ATP2A2, ZFP36, REST, HDAC9, STAC2, LOC153222, ZC3H12A, SLC25A37, IL17C, TRIB3, CEBPB, PTX3, TRIB3, KIAA1718, ZNF385, TTMP, MGC5618, CSF2, DUSP5, SLC25A25, ADAMTS1 , TFRC, SEC15L2, SCNN1A, HES1, LOC153222, C20orf72, ETS1, GDF15, KIAA1754, PACRG, PRSS22, TFRC, AQP2, STX3A, REM1, LIMK2, STC2, ISG20L1, BCL2A1, HES4, TJP3, SLC7A5, CEBPG, LILRB2 /// LILRB3, LOC389429, LLGL1 , KLF6, CSNK1 E, DUSP1 ,
ICAM1, PYY2, TPCN1, MFSD2, KIAA0690, SOD2, E2F7, GRIK5, PADM, HIP1R, IRAK2, EIF1, CEBPG, G0S2, KIAA0690, NEDD4L, MXD1.TXNDC5, PERLD1, HIF1A, RDH13, CTH, ETS2, FAH, CLK4, RIMS4, FLJ12355, LDLR, WARS, IFRD1, LRCH4, SLC1A4, FST, PPIF, ARHGEF2, PMAIP1, PBEF1, C14orf 159, ATF4, FLJ90119, TGFBR3, DDX21 , MUC8, TNFSF7, SPTLC2,
KBTBD8, CIAS1, KIAA0476, NFKB1, DES/// FAM48A, ICAM1, INHBA, MARS, C1 orf113, GRK6, TCF2, SLC7A1 , TNFRSF1 OB, IER2, TFRC, SOS2, CARS, HAB1 /// LOC442496, KLHL24, SQSTM1, GIT1, IRS2, SARS, TRIM25, JUN, YRDC, ETS1, CTH, ABL2, CD274, FZD8, EREG, PFDN2, TMEM63B, DYRK3, GARS, RASSF8, CAMTA2, HOXC8, AARS, TBX3, RRAGC, BCAP29,
TNFRSF10B, MCL1 , PALM2-AKAP2, TMEM22, ZNF548, PCBP2, SLC1A4, FSTL3, TA-PP2C, PIM3, MAFG, NOL1 , PEA15, ETS2, IQCG, BEX2, C12orf39, LDLR, VEGF, CCNL1 , LOC440069, SNAPC1 , SLC27A5, MARS, ICAM1, AXUD1.KCNIP2, LOC146177/// FLJ40941, GOT1 , C11 orf 17 /// NUAK2, TTBK1, SNCA, NFRKB, GRB2, ADAMTS1 , PALM2-AKAP2, CCNL1 , JUN, SPRY4,
SLC38A1, E2F7, MGC27165, IDS, SLC7A1 , FLJ11021 , KCNK5, GYPC, MYC, CACNG6, PHLDA1, INSIG1, FLJ13448, LOC202051 , GALNT10, TFEB, ABL2, MTHFD2, SERPINB8,TNPO2, MCF2L, SERTAD1, SPRY2, PHLDA1, SLC7A11 , CXCL5, COMT, GTPBP2, RAB4B, ATXN7, 0TUD5, VEGF, ADAMTS6, YARS, NAV3, SAMD4, C17orf32, SHMT2, CHIC2, 0RA0V1, CREM, ALS2CL,
AMIG02, MGC19764, SFPQ, MGC11257, PLEKHM1, MMP28, SLC4A5, DNAJB9, ERRFM, FZD8, MGC4399, GUCA1B, ATP13A3, KIAA0020, XBP1, ATP6V1B2, IGLV3-25, CHD4, GRB10, FLJ40089, CLDN19, NPC1, RIPK2, PLEKHF2, LOC283314, RSP03, GSK3A, ANKRD11 , WARS, GNPDA1, IGKC/// IGKV1-5, RCL1, C9orf91 , TIMM10, SLC1A4, 0DC1, SLC38A1 , EIF4EBP1,
ZNF267, YRDC, CHD4, PER1, NFKBIE, INSIG1, FLJ11259, NC0A3, TNP02, BAZ2A, SLC1A5, KRTAP2-1 , PDE4DIP, EPB41L3, CITED2, JTV1 , TA-PP2C, METRNL, BCL10, C1orf19, PTP4A1 , PDE4DIP, CPSF1, TIMM10, IGSF21, TM4SF1, CDK6, BANP, PHLDA1, PAPPA2, MGC17337, TNFRSF19L, ADAMTS10, CBFB, HERPUD1, RTTN, FLJ13855, SYNPO, NAV2, FLJ34208,
DDX10, C6orf66, LOC56902, CHMP1B, HIPK2, H3F3B, NAV3, ELL2, NSUN5C, PLAUR, UCK2, PBEF1, TP53BP2, TEX10, B3GNT5, USP36, INHBA, SPIRE1, CSTB, NSUN5C, TFPI2; or wherein the expression of the following genes is inhibited: FN1, ITGAV, LAMA4, LAMB2, LAMC1, LAMB1, LAMB1, LAMA4, LAMA5, LAMC1, LAMA2, LAMB1, LAMA3, SCAMP1 , TICAM2, SCAMP1, TICAM2, SCAMP1 , SCAMP1 , CAMK2B, DL1 ,
ICAMS5CEECAMIJCAMS5SCAMPI, CAMK1G, CAMSAP1, MCAM, CAMTA1, CKN1, ALCAM, DCAMKL2, CEACAM3, CAMK2D, CAMK2B, SCAMP5, CAMK4, NCAM1, CAMK2G, MYH9, MYH10, MYO1 D, MYO5A, MYLK, MY06, MYO5A, MYO1C, MYLK, MY06, MYLC2PL, MYO10, MY06, TPM3, MYO1C, BECN1, MYO1E, TPM3, M-RIP, MYO1B, MYO10, MYO5A, M-RIP, MYO10, MYL6,
MYOHD1, BECN1, TPM4, MYLK, MYH10, MYOHD1, LOC221875, LOC402643, MYO15B, LOC129285, MYH11, MYO1B, MYO1C, MYO9B, CDH13, CTNNAL1, CDH13, CDH12, CTNNB1, CDH5, CTNND1, CDH2, CTNNA1, CDH2, PCDHB16, CTNNA1, CELSR2, PCDHB6, PCDHB7, CTNND2, PCDHGC3, PCDHGB4, PCDHGA8, PCDHGA12, PCDHGC5 , PCDHGC4 , PCDHGB7,
PCDHGB6, PCDHGB5, PCDHGB3, PCDHGB2, PCDHGB1, PCDHGA11, PCDHGA10 , PCDHGA9, PCDHGA7, PCDHGA6, PCDHGA5, PCDHGA4 , PCDHGA3 , PCDHGA2, PCDHGA1, CTNND1, CDH23, PCDHB12, PCDHB10, PCDH18, CDH20, PCDH9, PCDHGA12, PCDHGA11, PCDHGA10, PCDHGA6, PCDHGA5, PCDHGA3, PCDH7, CDH18, CDH6, CCBE1 , COL10A1 , COL12A1 ,
COL13A1, COL18A1, COL1A1, COL21A1, COL4A1 , COL4A2, COL4A5, COL4A6, COL5A1 , COL5A2, COL6A1 , COL6A2, COL6A3, COL9A1 , MMP9, P4HA1, P4HA2, P4HB, PCOLCE, PCOLCE2, PCOTH, PL0D1, PL0D2, PL0D3, CIB1, ILK, ITGA2, ITGA3, ITGA4, ITGA6, ITGAV, ITGB1, ITGB1BP1, ITGB2, ITGB5, ITGBL1 , TNC, EMILIN1 , ICAM1 , HSPG2, HPSE, HS2ST1 ,SDC2,
EP09721583A 2008-02-15 2009-02-13 Blocking the metastasis of cancer cells and the uses of new compounds thereof Withdrawn EP2254899A4 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
PCT/US2008/002086 WO2008133766A1 (en) 2007-02-16 2008-02-15 Blocking the migration or metastasis of cancer cells by affecting adhesion proteins and the uses of new compounds thereof
US3827708P 2008-03-20 2008-03-20
US5430808P 2008-05-19 2008-05-19
US12/344,682 US8841265B2 (en) 2003-10-09 2008-12-29 Composition comprising triterpene saponins and compounds with angeloyl functional group, methods for preparing same and uses thereof
PCT/US2009/034115 WO2009117196A1 (en) 2008-02-15 2009-02-13 Blocking the metastasis of cancer cells and the uses of new compounds thereof

Publications (2)

Publication Number Publication Date
EP2254899A1 true EP2254899A1 (en) 2010-12-01
EP2254899A4 EP2254899A4 (en) 2012-05-30

Family

ID=41091216

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09721583A Withdrawn EP2254899A4 (en) 2008-02-15 2009-02-13 Blocking the metastasis of cancer cells and the uses of new compounds thereof

Country Status (5)

Country Link
EP (1) EP2254899A4 (en)
CN (1) CN101998964B (en)
AU (1) AU2009226063A1 (en)
CA (1) CA2715501A1 (en)
WO (1) WO2009117196A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2484990A2 (en) 2010-10-07 2012-08-08 Luke Chamberlain A boiler assembly comprising a removable boiler unit
CN102851388A (en) * 2012-09-29 2013-01-02 山东大学齐鲁医院 Molecular marker for identifying susceptibility of ovarian cancer, kit and identifying method thereof
CN102864234A (en) * 2012-09-29 2013-01-09 山东大学齐鲁医院 Molecular marker for identifying susceptibility of ovarian cancer

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201136604A (en) * 2009-12-14 2011-11-01 Oncotherapy Science Inc TMEM22 peptides and vaccines including the same
CN115197293A (en) * 2010-07-16 2022-10-18 太平洋艾瑞有限公司 Novel compounds useful for the treatment of cancer and other diseases
KR101873607B1 (en) * 2010-07-16 2018-07-02 퍼시픽 애로우 리미티드 New compounds for treating cancer and other diseases
WO2013061328A2 (en) 2011-10-27 2013-05-02 Yeda Research And Development Co. Ltd. Method of treating cancer
CN104062432B (en) * 2013-03-22 2016-01-20 中山大学 The application of ang2 in preparation ECTC vascular group diagnosing cancer of liver reagent and medicine
US10987369B2 (en) 2014-04-10 2021-04-27 Academia Sinica Pdia4 protein as a target for diagnosis, monitoring and treatment of diabetes
CN103992236B (en) * 2014-04-29 2016-05-18 于非 A kind of New Target tropism antineoplastic and preparation method thereof and application
CN105467127B (en) * 2015-03-23 2017-06-06 复旦大学 The application process and its detection kit of a kind of human colon carcinoma protein markers COL6A3
CN109803978B (en) * 2016-09-07 2022-07-01 武汉华大吉诺因生物科技有限公司 Polypeptide and application thereof
WO2018219342A1 (en) * 2017-06-01 2018-12-06 立森印迹诊断技术有限公司 Imprinted gene graded model and diagnostic method and application
CN107312855B (en) * 2017-07-24 2020-12-25 青岛泱深生物医药有限公司 Gene related to laryngeal squamous cell carcinoma and application thereof
CN107543926A (en) * 2017-08-25 2018-01-05 复旦大学附属金山医院 A kind of application of new Diagnosis of Breast invasive ductal carcinoma
TW201919644A (en) * 2017-09-29 2019-06-01 台灣浩鼎生技股份有限公司 Method for treating leukemia
CN108103194B (en) * 2017-12-27 2021-05-18 中国医学科学院药用植物研究所 Target for inhibiting gastric cancer cell MGC-803 and application thereof
CN108220421B (en) * 2018-01-05 2019-04-05 智海基因科技南京有限公司 The diagnostic kit and related gene of oneself immunity hepatitis are preparing the application in the kit
CN110272997B (en) * 2018-03-14 2023-04-28 华中科技大学同济医学院附属同济医院 Application of C/EBP beta gene or protein
CN108728539A (en) * 2018-06-05 2018-11-02 浙江大学 A kind of application of HoxD10 genes in gastric cancer
CN108840923B (en) * 2018-06-22 2021-08-03 上海交通大学医学院附属仁济医院 Polypeptide targeting PD-L1 and application thereof
CN108642181B (en) * 2018-06-25 2021-09-10 遵义医科大学附属医院 Application of long-chain non-coding RNA SLC25A25-AS1
CN111217900A (en) * 2018-11-27 2020-06-02 上海交通大学 Transcription regulation factor for angiogenesis and application thereof
CN111289749B (en) * 2018-12-10 2023-04-14 北京蛋白质组研究中心 Application of C-type 1-type Niemann-Pickerin detection substance in preparation of hepatocellular carcinoma screening product
CN109402119A (en) * 2018-12-11 2019-03-01 宁夏医科大学总医院 A kind of circular rna hsa_circTGFBI_001 and its specificity amplification primer and application
CN109402125A (en) * 2018-12-11 2019-03-01 宁夏医科大学总医院 A kind of circular rna hsa_circTGFBI_007 and its specificity amplification primer and application
CN110672848A (en) * 2019-10-25 2020-01-10 四川大学华西医院 Application of APEH autoantibody detection reagent in preparation of lung cancer screening kit
CN111153956B (en) * 2019-12-06 2020-12-11 江苏一片叶高新科技有限公司 Tea saponin compound, preparation method and application
CN110938630B (en) * 2019-12-20 2023-07-28 中国人民解放军第四军医大学 Application of human B3GNT5 gene and related products
CN111166769B (en) * 2020-03-18 2022-01-11 山东大学 Application of dermal fibroblast in treating melanoma
CN111979328B (en) * 2020-09-07 2022-10-14 复旦大学附属中山医院 Biomarker for predicting liver cancer lung metastasis and application thereof
CN112011620B (en) * 2020-09-23 2021-04-27 山东大学第二医院 Application of circ-SLC38A1 as target in medicine for inhibiting bladder cancer cells
CN112481373B (en) * 2020-12-21 2022-01-28 华北理工大学 circRNA detection kit for auxiliary diagnosis of autism
CN114015689B (en) * 2021-10-26 2024-06-07 江苏大学 ShRNA sequence for specifically inhibiting GOS2 gene expression and application thereof
CN114984187B (en) * 2022-07-20 2024-07-26 南通大学 Application of transmembrane protein Tmem63b in preparation of medicine for treating peripheral nerve demyelinating diseases

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000038700A1 (en) * 1998-12-22 2000-07-06 Janssen Pharmaceutica N.V. Antiprotozoal saponins
US20070161580A1 (en) * 2003-10-09 2007-07-12 Chan Pui-Kwong Anti-Tumor Compounds With Angeloyl Groups

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000038700A1 (en) * 1998-12-22 2000-07-06 Janssen Pharmaceutica N.V. Antiprotozoal saponins
US20040138151A1 (en) * 1998-12-22 2004-07-15 Janssen Pharmaceutica N.V. Antiprotozoal saponins
US20070161580A1 (en) * 2003-10-09 2007-07-12 Chan Pui-Kwong Anti-Tumor Compounds With Angeloyl Groups

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DATABASE CA [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; GERMONPREZ, NILS ET AL: "Antileishmanial saponins from Maesa", XP002673641, retrieved from STN Database accession no. 2003:888935 & GERMONPREZ, NILS ET AL: "Antileishmanial saponins from Maesa", TAP CHI HOA HOC , 41(SPEC.), 125-130 CODEN: TCHHDC; ISSN: 0378-2336, 2003, *
GERMONPREZ NILS ET AL: "In vitro and in vivo anti-leishmanial activity of triterpenoid saponins isolated from Maesa balansae and some chemical derivatives", JOURNAL OF MEDICINAL CHEMISTRY, vol. 48, no. 1, 13 January 2005 (2005-01-13), pages 32-37, XP002672062, ISSN: 0022-2623 *
LOUIS MAES ET AL: "In Vitro and In Vivo Activities of a Triterpenoid Saponin Extract (PX-6518) from the Plant Maesa balansae against Visceral Leishmania Species", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, AMERICAN SOCIETY FOR MICROBIOLOGY, WASHINGTON, DC, US, vol. 48, no. 1, 1 January 2004 (2004-01-01), pages 130-136, XP008140635, ISSN: 0066-4804, DOI: 10.1128/AAC.48.1.130-136.2004 *
LU Y ET AL: "Triterpenoid saponins from the roots of tea plant (Camellia sinensis var. assamica)", PHYTOCHEMISTRY, PERGAMON PRESS, GB, vol. 53, no. 8, 1 April 2000 (2000-04-01), pages 941-946, XP027262251, ISSN: 0031-9422 [retrieved on 2000-04-01] *
ODA K ET AL: "Adjuvant and Haemolytic Activities of 47 Saponins Derived from Medicinal and Food Plants", BIOLOGICAL CHEMISTRY, WALTER DE GRUYTER GMBH & CO, BERLIN, DE, vol. 381, 1 January 2000 (2000-01-01), pages 67-74, XP003005996, ISSN: 1431-6730, DOI: 10.1515/BC.2000.009 *
See also references of WO2009117196A1 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2484990A2 (en) 2010-10-07 2012-08-08 Luke Chamberlain A boiler assembly comprising a removable boiler unit
CN102851388A (en) * 2012-09-29 2013-01-02 山东大学齐鲁医院 Molecular marker for identifying susceptibility of ovarian cancer, kit and identifying method thereof
CN102864234A (en) * 2012-09-29 2013-01-09 山东大学齐鲁医院 Molecular marker for identifying susceptibility of ovarian cancer
CN102851388B (en) * 2012-09-29 2014-03-19 山东大学齐鲁医院 Molecular marker for identifying susceptibility of ovarian cancer, kit and identifying method thereof

Also Published As

Publication number Publication date
WO2009117196A1 (en) 2009-09-24
CN101998964A (en) 2011-03-30
AU2009226063A1 (en) 2009-09-24
WO2009117196A8 (en) 2009-11-26
CA2715501A1 (en) 2009-09-24
EP2254899A4 (en) 2012-05-30
CN101998964B (en) 2015-05-06

Similar Documents

Publication Publication Date Title
US8586719B2 (en) Triterpenes for modulating gene expression and cell membrane, and as antiprotozoal agents
EP2254899A1 (en) Blocking the metastasis of cancer cells and the uses of new compounds thereof
EP2121715A1 (en) Blocking the migration or metastasis of cancer cells by affecting adhesion proteins and the uses of new compounds thereof
US8735558B2 (en) Blocking the migration or metastasis of cancer cells by affecting adhesion proteins and the uses of new compounds thereof
JP2010519219A5 (en)
Yoshikawa et al. Medicinal flowers. III. Marigold.(1): hypoglycemic, gastric emptying inhibitory, and gastroprotective principles and new oleanane-type triterpene oligoglycosides, calendasaponins A, B, C, and D, from Egyptian Calendula officinalis
Myszka et al. Synthesis and induction of apoptosis in B cell chronic leukemia by diosgenyl 2-amino-2-deoxy-β-D-glucopyranoside hydrochloride and its derivatives
EP2473040A1 (en) Inhibiting the invasion and metastasis of cancer cells
Ohtsuki et al. New chlorogenin hexasaccharide isolated from Agave fourcroydes with cytotoxic and cell cycle inhibitory activities
US20120142620A1 (en) Core 2 glcnac-t inhibitors
JP2023126878A5 (en)
Shirahata et al. Syntheses and mucosal adjuvant activity of simplified oleanolic acid saponins possessing cinnamoyl ester
JP2004051641A (en) HEDERAGENIN 3-O-alpha-L-RHAMNOPYRAMOSYL(1->2)-[beta-D-GLUCOPYRANOSYL(1->4)]-alpha-L-ARABINOPYRANOSIDE ANDUSE OF DRIED PASQUEFLOWER ROOT EXTRACT CONTAINING THE SAME AS SOLID TUMOR DRUG
Grynkiewicz et al. Synthetic analogs of natural glycosides in drug discovery and development
Zeng et al. Synthesis and anti-cancer activity of a glycosyl library of N N-acetylglucosamine-bearing oleanolic acid
AU2015200182A1 (en) Blocking the metastasis of cancer cells and the uses of new compounds as antiprotozoal agents
Chen et al. New Dammarane Monodesmosides from the Acidic Deglycosylation of Notoginseng‐Leaf Saponins
Noté et al. Pro-apoptotic activity of new triterpenoid saponins from the roots of Albizia adianthifolia (Schumach.) W. Wight
Popov et al. Aphelasteroside F, a new asterosaponin from the far eastern starfish Aphelasterias japonica
Gu et al. Synthesis of Leonosides E and F derived from Leonurus japonicas Houtt
US20220024961A1 (en) Methods and compounds for modulating the secretion or expression of adhesion proteins or angiopoietins of cells
Ikeda et al. Chemical trans-glycosylation of bioactive glycolinkage: synthesis of an α-lycotetraosyl cholesterol
Schimmel et al. Synthesis of saponins with cholestanol, cholesterol, and friedelanol as aglycones
JP3282583B2 (en) Adjuvant composition, vaccine using the same and novel saponin
CZ200851A3 (en) Carbohydrate derivatives of lupane, their use and pharmaceutical preparations containing these derivatives

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20100906

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1150611

Country of ref document: HK

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/704 20060101ALI20120412BHEP

Ipc: A61K 31/7048 20060101ALI20120412BHEP

Ipc: A61P 33/02 20060101ALI20120412BHEP

Ipc: C07H 15/24 20060101AFI20120412BHEP

Ipc: C07H 15/00 20060101ALI20120412BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20120426

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20121127

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1150611

Country of ref document: HK