Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-TUMOR COMPOUNDS FOR INHIBITING CANCER GROWTH
Document Type and Number:
WIPO Patent Application WO/2008/028060
Kind Code:
A2
Abstract:
This invention provides a method for treating cancer by blocking the migration, metastasis of cancer cells, growth of cancers 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 provides uses of compositions comprising a triterpenoidal saponin, triterpenoid, triterpenoidal compound or sapongenin, comprising at least two side groups selected from the group consisting of angeloyl groups, tigloyl groups and senecioyl groups, wherein the side groups are attached to carbon 21, 22 or/and 28 of triterpenoidal sapogenin, triterpenoid, triterpenoidal compound or other sapongenin backbones.

Inventors:
CHAN PUI-KWONG (US)
MAK MAY SUNG (CN)
Application Number:
PCT/US2007/077273
Publication Date:
March 06, 2008
Filing Date:
August 30, 2007
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PACIFIC ARROW LTD (CN)
CHAN PUI-KWONG (US)
MAK MAY SUNG (CN)
International Classes:
A61K31/704; C07H15/24
Domestic Patent References:
WO2003037200A22003-05-08
Foreign References:
US6616943B22003-09-09
Other References:
See references of EP 2061798A4
Attorney, Agent or Firm:
CHAN, Albert, Wai-Kit (PLLCWorld Plaza, Suite 604,141-07 20th Avenu, Whitestone NY, US)
Download PDF:
Claims:

What is claimed is:

1. A use of a composition for the preparation of medicament to inhibite cancer growth, wherein the cancer is selected from the group consisting of 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, and wherein the composition comprises a compound having a formula:

named (1 B), or a salt, ester, metabolite or derivative thereof, wherein R1 comprises a group selected from hydrogen, angeloyl, tigloyl, senecioyl, alkyl, dibenzoyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl or derivatives thereof; R2 comprises a group selected from hydrogen, angeloyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl or derivative thereof; R4 comprises CH 2 OR6 or COOR6, wherein R6 is selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocyclic, heteroraryl or an acid with carbon 2-6, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl or derivative thereof; R3 is H or OH; R8 is OH; R5 comprises a hydrogen or sugar moiety, wherein the sugar moiety comprises one or more sugars including but 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; wherein 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, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl particularly CH 3 , CH 2 OH.

2. According to claim 1 , the composition comprising a compound wherein at least two of R1 , R2, and R4 comprise groups selected from angeloyl, tigloyl, senecioyl, alkyl,

benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivative thereof; or at least one of R1 , R2, and R4 comprises a sugar moiety comprising at least two groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocyclic, heteroraryl or an acid with carbon 2-6, or a derivative thereof.

3. According to claim 1 , the composition comprising a compound, wherein R4 comprises CH 2 OR6 wherein R6 is H; wherein R1 and R2 independently comprise an angeloyl group, or at least two of R1 , R2 and R6 comprises angeloyl or at least one of R1 , R2 or R6 comprises a sugar moiety with two angeloyls I; wherein R5 comprises a sugar moiety wherein the sugar moiety is comprised glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid or derivative thereof, or the combination thereof.

4. According to Claim 1 , the composition comprising a compound selected from the following: a) A compound comprising 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) A compound comprising structure Xanifolia (Y1 ),

chemical name: 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl (1→3)-β-D-glucuronopyranosyl-21-O-(3,4-diangeloyl)-α- L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28-pentahydroxyolean-12-ene; c) A compound comprising 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) A compound comprising structure Xanifolia (Y8),

or chemical name: 3-O-[/?-glucopyranosyl (1→2)]-α-arabinofuranosyl (1^3)-/?-glucuronopyranosyl-21 , 22-O-diangeloyl-3/?, 16«, 21/?, 22«, 24/?, 28-hexahydroxyolean-12-ene; e) A compound comprising structure Xanifolia (Y9),

or chemical name: 3-0-[β- galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21-O-(3,4- diangeloyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean- 12-ene; and f) A compound comprising structure Xanifolia (Y10),

or chemical name:

3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-O-diangeloyl-3# 16«, 21$ 22«, 28-pentahydroxyolean-12-ene. g) A compound comprising 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-O-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12- ene, h) A compound comprising structure Xanifolia (X),

or chemical name: 3-0-{[β-D- galactopyranosyl (1→2)]-[«-L-arabinofuranosyl (1→3)]-/?-D-glucuronopyranoside butyl ester}-21-O-acetyl-22-O-angeloyl- 3y?,16«,21#22«,28-pentahydroxyolean-12-ene. i) A compound comprising Compound (Y7),

or chemical name: 3-O-[β-D- galactopyranosyl-(1 →2)]-α-L-arabinofuranosyl-(1 →3)-j8-D-glucuronopyranosyl-21 -O- angeloyl-28-O-2-methylbutanoyl-3i8, 15 a, 16α, 21/3, 22α, 28-hexahydroxyolean-12- ene

5. According to Claim 1 , the composition comprising a compound selected from the following: a) 3-O-[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;

b) 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl (1→3)-β-D- glucuronopyranosyl-21-O-(3,4-diangeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28-pentahydroxyolean-12-ene; c) 3-O-[β-D-glucopyranosyl-(1→2)]-α-L-arabinofuranosyl (1 →3)- β-D- glucuronopyranosyl-21 ,22-O-diangeloyl-3/?, 15«, 16«, 21/?, 22«, 24/?, 28- heptahydroxyolean-12-ene; d) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl- 21 , 22-O-diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene; e) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl- 21-O-(3,4-diangeloyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28- pentahydroxyolean-12-ene; f) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-O-diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene; g) 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl (1 →3)-β-D-glucuronopyranosyl-21 ,22-O-dibenzoyl-3β, 15α,

16α, 21 β, 22α, 28-hexahydroxyolean-12-ene; h)3-O-[β-D-galactopyranosyl(1 →2)]-α-L arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21-O-(3,4- dibenzoyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α,

21 β,22α, 28-pentahydroxyolean-12-ene; i)3-O-[β-D-glucopyranosyl-(1 →2)]-α-L-arabinofuranosyl (1 →3)-(-D-glucuronopyranosyl

-21 ,22-0- dibenzoyl -3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12-ene; j) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21 ,

22-0- dibenzoyl -3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene; k) 3-O-[/?-galactopyranosyl (1 →2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl- 21-O-(3,4-dibenzoyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28- pentahydroxyolean-12-ene;

I) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl -

21 , 22-0- dibenzoyl -3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene; m) 3-O-[β-D-galactopyranosyl (1→2)]-β-D-xyopyranosyl (1→3)-β-D- glucuronopyranosyl-21 , 22-O-dibenzoyl-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene; n) 3-O-[β-D-galactopyranosyl(1→2)]- β-D-xyopyranosyl (1→3)-β-D- glucuronopyranosyl-21-O-(3,4-dibenzoyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α,

21 β, 22α, 28-pentahydroxyolean-12-ene; o) 3-O-[β-D-glucopyranosyl-(1→2)]-β-D-xyopyranosyl (1→3)- /?-D-glucuronopyranosyl-

21 , 22-0- dibenzoyl -3/?, 15«, 16«, 21 R 22«, 24/?, 28-heptahydroxyolean-12-ene;

p) 3-O-[/?-D-galactopyranosyl (1 →2)]- β- D-xyopyranosyl (1 →3)-^- D- glucuronopyranosyl-21 , 22-0- dibenzoyl -2>β, 16«, 21/?, 22«, 28-pentahydroxyolean-

12-ene; q) 3-O-[/?-galactopyranosyl (1 →2)]- β- xyopyranosyl (1 →3)-/?-glucuronopyranosyl-21- 0-(3A- dibenzoyl)-«-rhamnopyranosyl-28-O-acetyl-3y?, 16«, 2^β, 22«, 28- pentahydroxyolean-12-ene; r) 3-O-[/?-galactopyranosyl (1 →2)]-β- xyopyranosyl (1 →3)-/?-glucuronopyranosyl-21 ,

22-0- dibenzoyl -3β, 16«, 21$ 22«, 28-pentahydroxyolean-12-ene; s) 3-O-[β-D-galactopyranosyl (1 →2)] - β- D-xyopyranosyl (1 →3)-β-D- glucuronopyranosyl-21 , 22-O-diangeloyl-3β, 15α,16α, 21 β, 22α, 28- hexahydroxyolean-12-ene; t) 3-O-[β-D-galactopyranosyl (1 →2)] - β- D-xyopyranosyl (1 →3)-β-D- glucuronopyranosyl-21-O-(3, 4-diangeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-

3β, 16α, 21 β, 22α, 28-pentahydroxyolean-12-ene; u) 3-O-[β-D-glucopyranosyl-(1 →2)] - β- D-xyopyranosyl (1 →3)- β-D- glucuronopyranosyl-21 , 22-O-diangeloyl-3y?, 15«, 16«, 21$ 22«, 2λβ, 28- heptahydroxyolean-12-ene; v) 3-O-[/?-galactopyranosyl (1 ^2)] - β- D-xyopyranosyl (1 →3)-y#-glucuronopyranosyl-

21 , 22-O-diangeloyl-3y?, 16«, 2λβ, 22«, 28-pentahydroxyolean-12-ene; w) 3-O-[/?-galactopyranosyl (1 →2)] - β- D-xyopyranosyl (1 →3)-/?-glucuronopyranosyl-

21-0- (3, 4-diangeloyl)-«- rhamnopyranosyl-28-O-acetyl-3y?, 16«, 2λβ, 22«, 28- pentahydroxyolean-12-ene; x) 3-O-[y?-D-galactopyranosyl (1 →2)] - β- D-xyopyranosyl (1 →3)-y?-D- glucuronopyranosyl-21 , 22-O-diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean- 12-ene; and

Y) A compound comprising chemical name: 3-O-[/?-galactopyranosyl (1 →2)]-«- arabinofuranosyl (λ →3)-β- glucuronopyranosyl-21 -O-(3, 4-diangeloyl)-«-fucopyranosyl

-28-O-acetyl-3y?, 16«, 2λβ, 22«, 28-pentahydroxyolean-12-ene

6. The use of any one of claims 1-5, wherein the medicament is effective for extending the life span of a cancer-bearing mammal, or reducing the size of tumor in a mammal.

7. The use of any one of claims 1-6, wherein the composition comprises a compound comprising:

or

wherein R1 and R2 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative thereof; or particularly wherein the R1 and R2 comprise angeloyl groups; wherein the compound further comprises a sugar moiety, wherein the sugar moiety comprise 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; or wherein the compound comprise:

wherein at least two of R1 , R2 and R3 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative thereof; particularly, wherein at least two of the R1 , R2 and R3 comprise angeloyl groups, or wherein at least one of R1 , R2 and R3 comprise a sugar moiety which comprises two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6, particularly comprising two angeloyl groups, or wherein the compound comprise:

wherein R1 , R2 or R3 comprise angeloyl groups, tigloyl groups, senecioyl groups or acetyl group, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocyclic, heteroraryl, or acid with 2 to 5 carbon or their combinations, preferable wherein at least two of the R1 , R2 and R3 comprise angeloyl

groups; or wherein at least two of R1 , R2 and R3 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocyclic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6; or wherein 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, heterocyclic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6; wherein R5 comprises a sugar moiety, wherein the sugar moiety is comprised glucose, galactose, arabinose, alduronic acid, glucuronic acid, galacturonic acid or derivative thereof, or the combination thereof.

8 The use of any one of claims 1-7, wherein a medicament is intravenous injection or intravenous drip; wherein a medicament is intravenous drip: 0.05-0.2mg/kg medicine dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution; or wherein a medicament is intravenous injection: 0.05-0.2mg/kg/day medicine dissolved in 10-2OmI of 10% glucose solution or of 0.9% NaCI solution; or wherein a medicament is intravenous drip: 0.1-0.2mg/kg/day medicine dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution; or wherein a medicament is intravenous injection: 0.1-0.2mg/kg/day medicine dissolved in 10-2OmI of 10% glucose solution or of 0.9% NaCI solution; or wherein a medicament is intraperitoneal (I. P.): 2.5mg/kg/day medicine dissolved in 10% glucose solution or of 0.9% NaCI solution; or wherein a medicament is administered orally wherein the dosage of mammal is 1-10mg/Kg. ; or wherein a medicament is administered orally wherein the dosage is 10-30mg/Kg; or wherein a medicament is administered orally wherein the dosage is 30-60mg/Kg; or wherein a medicament is administered orally wherein the dosage is 60-90mg/Kg ; or wherein a medicament is administered by intravenous injection or intravenous drip wherein the dosage of mammal is 0.01- 0.1 mg/Kg, or wherein the dosage is 0.1- 0.2mg/Kg, or wherein the dosage is 0.2 - 0.4mg/Kg, or wherein the dosage is 0.4 - 0.6 mg/Kg; or a medicament is administered by intraperitoneal (I. P.) wherein the dosage of mammal is 1-3mg/Kg, or wherein the dosage is 3-5mg/Kg, or wherein the dosage is 4-6mg/Kg, or wherein the dosage is 6-10mg/Kg.

9. The use of any one of claims 1-8, wherein the cancer comprises cells overexpressing aquaporin.

10. The use of any one of claims 1-9, wherein the medicament is capable of interacting or regulating the protein on the surface of a cell or altering the functional properties of

intracellular membranes or regulating the fluid passage through the cell wall or softening the skin or improving the skin structure.

The use of any one of claims 1-8, wherein the medicament is also capable of 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, dysmenorrhea!, hemorrhoids, episiotomies, haemonhoids, peripheral oedema formation or postoperative swelling; for reducing symptoms of leg pain; for treating pruritis, lower leg volume, thrombosis, thromophlebitis; for preventing gastric ulcers antispasmotic, for antithrombotic, anti-oedematous or anti inflammatory treatment, for regulating the release of PGF2, antagonism to 5-HT and histamine; or for reducingcatabolism of tissue mucopolysaccharides, for AntiMS, antianeurysm, antiasthmatic, antibradykinic, anticapillarihemorrhagic, anticephalagic, anticervicobrachialgic, antieclamptic, antiedemic, antiencaphalitic, antiepiglottitic, antiexudative, antiflu, antifracture, antigingivitic, antihematomic, antiherpetic, antihistaminic, antihydrathritic, antimeningitic, antioxidant, antiperiodontic, antiphlebitic, antipleuritic, antiraucedo, antirhinitic, antitonsilitic, antiulcer, antivaricose, antivertiginous, cancerostatic, corticosterogenic, diuretic, fungicide, hemolytic, hyaluronidase inhibitor, lymphagogue, natriuretic, pesticide, pituitary stimulant, thymolytic, vasoprotective, venotonic treatment, for curing enuresis, frequent micturition, urinary incontinence, for curing disease by mechanisms related to increasing hemolytic activities or blood circulation or reducing the formation of blood clots.

Description:

ANTI-TUMOR COMPOUNDS FOR INHIBITING CANCER GROWTH

This application claims benefit or priority of U.S. Serial Nos. 60/841 ,727, filed September 1 , 2006, 60/890,380, filed February 16, 2007, and International Application No. PCT/US2006/016158, filed April 27, 2006, which claims benefit or priority of the following applications: (1 ) U.S. Serial Nos. 11/289,142, filed November 28, 2005, and 11/267,523, filed November 4, 2005; (2) International Application No. PCT/US2005/031900, filed September 7, 2005 (which claims the priority of U.S. Serial Nos. 60/617,379, filed October 8, 2004, 60/613,811 , filed September 27, 2004, and 60/607,858, filed September 7, 2004); (3) U.S. Serial No. 11/131 ,551 , filed May 17, 2005; and (4) U.S. Serial No. 11/117,760, filed April 27, 2005. This application also is a Continuation-ln-Part of U.S. Serial No. 10/906,303, filed February 14, 2005, which is a Continuation-ln-Part of International Application No. PCT/US04/43465, filed December 23, 2004, which is a Continuation-ln-Part of International Application No. PCT/US04/33359, filed October 8, 2004, which claims the benefit of U.S. Serial Nos. 60/532,101 , filed December 23, 2003, and 60/509,851 , filed October 9, 2003. The contents of these preceding applications are hereby incorporated in their entireties by reference into this application.

FIELD OF THE INVENTION The compounds and compositions in this invention inhibit tumor or cancer growth.

BACKGROUND OF THE INVENTION

We have identified an herbal extract from Xanthoceras sorbifolia that inhibits cancer cell's growth. The active compounds were purified and their structures identified to be novel triterpenoid saponins. We named them as Xanifolia-Y and family.

Varicose veins are swollen and knotted veins that can occur in any part of the body, especially in the calf, inside leg or around the anus. Escin has been satisfactorily used for treating Varicose veins and chronic venous insufficiency for many years. Escin is a mixture of saponins found in the seed of the horse chestnut tree, Aesculus hippocastanum L, Hippocastanaceae. Escin is the major active ingredient prepared from Aesculus hippocastanum (Hippocastanaceae), the horse chestnut tree. In one controlled trial study, aescin was shown to be as effective as compression therapy as an alternative to medical treatment for CVI. The therapeutic benefit is well supported by a number of experimental investigations in different animal models. See Department of Pharmacological Sciences, University of Milano, Via Balzaretti 9, 20133 Milano, Italy. New saponin compounds with two angeloyls have been provided in International PCT Application No. PCT/US04/33359, filed October 8, 2004, and U.S. Serial No. 10/906,303. Yingjie Chen, Tadahiro Takeda and Yukio Ogihara reported four new saponin compounds that were isolated from the fruits of Xanthoceras sorbifolia Bunge in Chem. Pharm. Bull., 33(1 )127-134,1985; 33(3)1043-

1048,1985 and 33(4)1387-1394,1985. Other related studies on saponin compounds include: triterpenoid saponins and acylated prosapogenins from Harpullia austro-caledonica (Voutquenne et al. 2002); six triterpennoid saponins from Maesa laxiflora (Zhong et al. 1999); new triterpene saponin from Pittosporum viήdiflorum from the Madagascar rainforest (Young et al. 2002); anti-HIV-1 protease triterpenoid saponins from the seeds of Aesculus chinensis (Yang et al. 1999); triterpenoid saponins from the roots of Camellia sinensis var. assamica (Lu et al. 2000); new acylated triterpenoid saponins from Maesa laceceolata (Apers et al. 1999); isolation and structure elucidation of four new triterpenoid estersaponins from fruits of the Pittosporumtobira AIT (D'Acquarica et al. 2002) and method for the prevention and treatment of chronic venous insufficiency (U.S. patent 6,210,680). The contents of the above-mentioned references are hereby incorporated by reference.

This invention shows that the saponins with two angeloyl groups have a strong activity for inhibiting cancers cells, anti-angiogenisis, inhibiting chronic venous insufficiency (CVI), shrinking hemorrhoids, inhibiting post-operative edema, rheumatism and cancerous cell growth.

This invention discloses saponin compounds having the specific structures that are capable of inhibiting cancer or tumor cell growth.

Human cells are surrounded by aquatic environments. Aquaporins is a family of transmembrane water-channel transporting proteins that play a major role in trans-cellular and transepithelial water movement. This invention shows that the triterpene saponins with two angeloyls have stronger activity for inhibiting cancer cell growth by affecting membrane functions. In an embodiment they affect the aquaporin and permeability of cell membrane.

SUMMARY OF THE INVENTION

In accordance with these and other objects of the invention, a brief summary of the present invention is presented. Some simplifications and omissions may be made in the following summary, which is intended to highlight and introduce some aspects of the present invention, but not to limit its scope. Detailed descriptions of a preferred exemplary embodiment adequate to allow those of ordinary skill in the art to make and use the invention's concepts will follow in later sections.

This invention provides the uses of compounds comprising a triterpene or other sapongenin with two angeloyl groups, or at least two side groups selected from the following groups: angeloyl, tigloyl and/or senecioyl groups, wherein the side groups are attached to carbon

21 , 22 or/and 28 of triterpenoidal saponin, triterpenoid, triterpenoidal compounds or other sapongenin backbones. The methods of purification and determination of structures of the compounds are detailed in the International Application No. PCT/US05/31900, filed September 7, 2005, U.S. Serial No. 11/289142, filed November 28, 2005 and U.S. Serial

11/131551 , filed May 17, 2005, the contents of which are incorporated herein by reference.

We have identified an herbal extract from Xanthoceras sorbifolia that inhibits cancer cell's growth. The active compounds were purified and their structures identified to be a novel triterpenoid saponin. We named them as Xanifolia-Y and family. Details are in U.S. Serial No. 10/906,303 and Int'l App'l No. PCT/US2004/033359.

In vivo studies with Xanifolia-Y employing human ovarian carcinoma xenografts in mouse indicate that Xanifolia-Y is capable of extending the life span of animals bearing human tumors. These results show that it can be useful in treating cancers in mammal. In an embodiment it can be use in treating human cancers, preferably ovarian cancer.

Xanifolia-Y prolongs the life span of mice bearing of human tumor. It blocks the migration or metastasis of cancer cells. In an embodiment it affects adhesion proteins or interferes with the function of molecules on carcinoma cells or on the mesothelial cells. It inhibits the tumor growth in mammal. It is useful in cancer therapy. See Experiment 7, 8, 9, and 10.

This invention discloses saponin compounds having the specific structures are capable of inhibiting cancer or tumor cell growth and anti-angiogenesis.

This invention provides a method 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.

This invention relates to the mechanism of inhibiting cancer by regulating aquaporin in cancer cell and the interacting of aquaporin with compounds comprise a triterpene and two angeloyl groups. In an embodiment, the compound may be a saponin wherein comprises at least one angeloyl, preferable two angeloyl groups.

This invention relates to the aquaporin pathway that is influenced by saponins with angeloyl groups in inhibiting cancer.

DETAILED DESCRIPTION OF THE FIGURES

Figure 1. Structure of saponin comprising two angeloyl groups. Figure 2 A, 2 B. Structures of saponins Figure 3. Structures of saponins

Figure 4 A and B. Comparison of potency of compound Y (saponin with 2 angeloyl groups) and compound X (saponin with 1 angeloyl) in inhibiting growth of ovarian cancer cells. The IC50 for Compound Y is about 1.5μg/ml while 30 μg/ml for compound X. Figure 4 C. Inhibition of growth of skin cancer cells by the purified compound Y. The IC50 is 0.23 μg/ml.

Figure 4 D. Hemolytic activity of Xanifolia-Y, B-Escin, Xanifolia-X, ACH-Y and AKOH-Y Figure 5. A. Anticancer activities of Y, Y8, Y9 and Y10, determined by MTT assay on ovarian cancer cells. B. The purified compound Y1 and compound Y2 show inhibition of growth of ovarian cancer cells. C. Compound Y inhibits tumor growth (IC50=4 μg/ml). Compound X which has a similar structure to Y but with only one angeloyl group at C22, has less anticancer activity (IC50=6 μg/ml). Removal of sugars from Y (ACH-Y) but retaining the diangeloyl group retains 40% of the anticancer activity (IC50=9.5 μg/ml). However, removal of both angeloyl groups from Y (AKOH-Y) completely abolishes its anticancer activity (even at 120 μg/ml). The results indicate that diangeloyl groups in compound Ys are important for anti-tumor activity.

Figure 5 D Compare inhibition activity of Xanifolia-Y, B-Escin, Xanifolia-X and AKOH-Y Figure 6. Comparision of MTT and hemolytic activities of saponin compound and compound Ys. (A) and (B). Hemolytic activities; (C) and (D). MTT activities. Figure 7. Saponin compound Y, X, ACH-Y, AKOH-Y and B-Escin These compounds are purified and their structures were verified by NMR and MS. These compounds are then used for MTT test. Figure 8 A: NMR 1 H of YO. B: NMR 13C of YO Figure 9 A: TOCSY of YO. B: HMQC of YO Figure 10 A: HNBC of Y0. B: NOESY of Y0 Figure 11 Structure of YO

Figure 12-16 show Xanifolia Y1 inhibits Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma Ovarian cancer, Renal cancer, Prostate cancer and Breast cancer activities

Figure 17-21 show Xanifolia Y2 inhibits Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma Ovarian cancer, Renal cancer, Prostate cancer and Breast cancer activities

Figure 22 Animal study result shows Group A Mice - Implanted tumor and no drug, Died on day 19-22; Group B Mice - Implanted tumor and with drug, survived over 50 days; Group

C Mice - No tumor and with drug, survived over 50 days.

Figure 23 Animal study result shows Group A Mice implanted with tumor and no drug, all died within 24 days; Group D Mice implanted with tumor and were given drug 9 times from

4th day, all survived; Group E Mice implanted with tumor and were given drug 10 times from

10th day, half the number of mice survived.

Figure 24 Animal study shows that the tumor size is 45% smaller in mice with drug than the mice with no drug in 10 days period. Figure 25 Study apoptosis induced by Xanifolia-Y that apoptosis is a major form of cell death induced by Xanifolia-Y.

Figure 26EM study the effect of Xanifolia on membrane show that patches of pits were found in the membrane of Xanifolia-Y treated cells (Fig. 34B) but not in cells treated with the

DMSO (Fig. 34A) or AKOH-Y (Fig. 34C) controls. These pits have the size from 8OA to 500A (in diameter). The pits represent holes formed in the membrane. The pits are arranged in a characteristic pattern with smaller pits (8OA in diameter) located in the periphery and the bigger ones (500A in diameter) in the center. The bigger holes are resulted from fusing of the smaller holes (Fig. 34D). Membrane image of cells treated with

A: DMSO solvent control, 60 min (magnification: X60,000); B: Xanifolia-Y 5 uM, 60 min. (X60000); C: AKOH-Y, 20 uM, 60 min. (X60000); D: Xanifolia-Y 5 uM, 60 min. (X20000)

Figure 27 Inhibition effect of Xanifolia and Paclitaxel on cancer cell

Figure 28 Activities of Ys

Figure 29 Animal survival experiment

Figure 30 Y7 NMR profile, A: HNMR, B: HMBC Figure 31 Y7 NMR profile, A: HMQC, B: NOESY

Figure 32 Determination of Aquaporin

Figure 33 Compare the potency of Xanifolia Y in ovary and cervix cell.

Figure 34-36 show Xanifolia YO inhibits Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma Ovarian cancer, Renal cancer, Prostate cancer and Breast cancer activities

Figure 37-39 show Xanifolia Y9 inhibits Leukemia cancer, Lung cancer, Colon cancer, CNS cancer, Melanoma Ovarian cancer, Renal cancer, Prostate cancer and Breast cancer activities

DETAILED DESCRIPTION OF THE INVENTION

This invention provides the results of a program for screening the bioactive compounds from natural plants. 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. 11/289142, filed November 28, 2005, and U.S. Serial No. 11/131551 , filed May 17, 2005, the contents of which are incorporated herein by reference

This invention provides the uses of compositions comprising a triterpenoidal saponin. In an embodiment, the saponin has triterpenoid, triterpenoidal or other sapongenin, one or more sugar moieties and two angeloyl groups, or at least two side groups selected from the following groups: angeloyl groups, tigloyl groups or senecioyl groups, wherein the side groups are attached to the sapongenin backbone at carbon 21 and 22. In an embodiment, at least two of angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl attached to the side groups; wherein the sugar moiety in the saponin comprises at least one or more of 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.

This invention further provides a composition comprising the structures comprising at least two side groups selected from the following groups: angeloyl, tigloyl or senecioyl groups, wherein the side groups are attached to a triterpenoidal, triterpenoid, triterpenoidal or other sapongenin backbone. These compositions are obtainable from the above-identified plants or synthesis.

This invention provides a method of preparing the saponins, comprising the steps of:

(a) Extracting roots, kernels, leaves, bark, stem, husks, seeds, seed shells or fruits of the above plant, or combinations thereof with organic solvents such as ethanol or methanol to obtain an organic extract; (b) Collecting the organic extracts; (c) Refluxing the organic extract to obtain a second extract; (d) Removing the organic solvent from the second extract to obtain a third extract; (e) Drying and sterilizing the third extract to obtain a crude extract powder; (f) Fractionating the crude extract powder into fractions or components. Fractionation may be achieved by HPLC and FPLC chromatography with silica gel, C18 or other equivalent solid phase materials; (g) Monitoring the fractionating, if using HPLC or FPLC, the absorption wavelength at 207nm to 500nm may be used; (h) Identifying the

bioactive components of the crude extract; (i) Purifying one or more bioactive components of the crude extract with FPLC to obtain one or more fractions of the bioactive component; and (j) isolating the bioactive components with chromatographic techniques that employ preparative columns and HPLC.

In an embodiment, this invention provides the method of MTT Assay to test the bioactivities of the saponins or other compounds.

Cells. Human cancer cell lines were obtained from American Type Culture Collection:

HTB-9 (bladder), HeLa-S3 (cervix), DU 145 (prostate), H460 (lung), MCF-7 (breast), K562 (leukocytes), HCT116 (colon), HepG2 (liver), U2OS (bone), T98G (brain), SK-MEL-5 (Skin) and OVCAR-3 (ovary). The cells were grown in following culture media: HeLa-S3, DU 145, MCF-7, Hep-G2 and T98G are in MEN (Earle's salts); HTB-9, H460, K562 and OVCAR-3 in RPMI-1640; HCT-1 16 and U2OS in McCoy-5A. They are supplemented with 10% fetal calf serum, glutamine and antibiotics, and incubated in an incubator with 5% CO 2 humidified at 37 0 C.

MTT Assay. The procedure for MTT assay followed the method described by Carmichael et al.(1987) with modifications. The cells were seeded into a 96-well plate at concentration of 10,000/well for HTB-9, HeLa, H460, HCT116, T98G and OVCAR-3), 15,000/well for DU145, MCF-7, HepG2 and U2OS), and 40,000/well for K562 for 24 hours before drug- treatment. The cells were then exposed to the drugs for 48 hours (72 hours for HepG2 and U2OS, and 96 hours for MCF-7). After the drug-treatment, MTT (0.5 mg/mL) was added to cultures and incubated for an hour. The formazan (product of the reduction of tetrazolium by viable cells) formed and was dissolved with DMSO and the O. D. at 490nm, and was measured by an ELISA reader. The MTT level of the cells before drug-treatment was also measured (TO). The % cell-growth (%G) is calculated as: %G = (TD-TO / TC-TO) x 100(1 ), where TC or TD represents O. D. readings of control or drug-treated cells.

When TO > TD, then the cytotoxicity (LC) expressed as % of the control is calculated as: %LC = (TD-TO / TO) x 100(2).

This invention provides a composition that effectively reduced or inhibitied the cancer cell growth, wherein the cancer includes but is not limited to bladder cancer, bone cancer and ovary cancer.

This invention provides a composition comprising an effective amount of triterpenoidal saponins named as Xanifolia Y1 , Y2, Y, Y7, Y8, Y9, Y10, YO or their derivatives 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 and inhibiting tumor growth.

This invention provides a method of inhibiting cancer cell growth by affecting the aquaporin protein. This invention provides a method of inhibiting tumor growth in a subject comprising administering contracting an effective amount of compounds in this invention to the subject affecting or interacting the aquaporin protien at the surface of cancer cell. The compound comprises two angeloyl groups. In an embodiment the compound may be selected from formula (1 ), (1A), (1B), (1C) and (1 D). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment the compound(s) are selected from Xanifolia (YO, Y1 , Y2, Y, Y7, Y8, Y9, and Y10). In an embodiment the compound(s) are selected from Xanifolia (x), Escin or Aescin. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application.

This invention provides a method of inhibiting cancer cell growth by increasing the static charge of the cell, wherein increase water flow in the cell. In an embodiment the compounsopen the channel protein or ion gates of the cells. The charged molecules or ions pass cell membrane through channel protein and kill cancer cell.

As used herein, the term "inhibit" encompasses prevent, and killing of the said cancer or tumor cell

This invention provides a method interacting with aquaporin protein for regulating the water channel, modulating the secretion, regulating the water metabolism of body, reducing the amount of urine, reducing urinate times, treating enuresis, treating frequent urination. The method comprises administering contracting an effective amount of compounds to the subject affecting or interacting with the aquaporin protein at the surface of caner cell. The compound comprises two angeloyl groups. In an embodiment the compound may be selected from formula (1 ), (1A), (1 B) and (1 D). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment, the compound comprises a triterpene backbone, two acetyl groups with 2 or more carbon and sugar moiety. In an embodiment the compound(s) are selected from Xanifolia (YO, Y1 , Y2, Y, Y7, Y8, Y9, and Y10). In an embodiment the compound(s) are selected from Xanifolia (x), Escin or Aescin. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application.

This invention provides uses compound comprises a triterpene and angeloyl groups interacting with aquaporin protein for regulating the water channel, modulating the secretion, treating enuresis, inhibiting tumor growth, stopping cancer cell proliferate. In an embodiment, compound interacting with aquaporin protein for regulating the water channel, modulating the secretion, destroying the cancer cell.

This invention provides a composition interacting with aquaporin protein for regulating the water channel, modulating the secretion, treating enuresis, inhibiting tumor growth. A composition comprising an effective amount of the compound of any one of YO, Y1 , Y2, Y,Y7, Y8, Y9, Y10, or a salt, ester, metabolite or derivative thereof as a medicament 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.

The aquaporins (AQPs) are a family of homologous water channels expressed in many epithelial and endothelial cell types involved in fluid transport. The family of mammalian AQPs consists of 11 members, AQP0-10, each with a distinct tissue. Functional measurements indicate that mammalian AQPs 1 , 2, 4, 5, and 8 are probably water selective, whereas AQPs 3, 7, 9, and 10 also transport glycerol and other small solutes. They are expressed at part of membrane of cell. AQP1 protein is strongly expressed in most microvessel endothelia outside of the brain, as well as in endothelial cells in cornea, intestinal lacteals, and in other tissues

AQP 1 protein was strongly expressed in the membrane of microvessels and small vessels in all ovarian epithelial tumors, but less at the cytoplasm of tumor cells. In addition, AQP1 protein was also observed in the membrane of interstitial cells of ovarian carcinoma. Incorporated by reference of: The influence of aquaporin-1 and microvessel density on ovarian carcinogenesis and ascites formation, J. H. Yang et al., 2006 IGCS, International Journal of Gynecological Cancer 16 (suppl. 1 )

Structural determinants of water permeation through aquaporin-1 , by Kazuyoshi Murata, et al., Nature, Vol. 407, October 5, 2000

The distribution amount of AQPs 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 are varied at membrane of different cells. In different tumor cell, certain type of Aquaporin proteins is over-expressed. An increasing number of disturbances have been found associated to abnormal function of these proteins. The compounds Xanifolia can interact with the aquaporin and inhibit the tumor cell growth. We used Western blot analysis and identified expression of aquaporin in

cell lines. Detail of Xanifolia in PCT/US2005/031900, filed September 7, 2005, and U.S. Serial 11/131551 , filed May 17, 2005.

A Western blot is a common method in molecular biology/biochemistry/immunogenetics to detect protein in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate proteins by mass. The proteins are then transferred out of the gel and onto a membrane, where they are "probed" using antibodies specific to the aquaporin protein. As a result, we can examine the amount of aquaporin protein in a given sample and compare levels between several groups. Other techniques which allow detection of proteins in tissues (immunohistochemistry) and cells (immunoctochemistry) are used. Other methods such as Bradford protien assay, UV spectroscopy, Biuret protein assay, Lowry protien assay, Bicinchonic acid protein assay may also be used.

There are many publications about the studies of the aquaporin as a maker for cancer cells but none of them mention the regulating or affecting the aquaporin as method to facilitate the blockage, inhibition or destroying the cancer cells.

This invention describes a method of destroying cancer cell or inhibiting the cancer cell proliferates by regulating or affecting the aquaporin. In an embodiment, the saponin with two angeloyl groups can interacts with the aquaporin in order inhibiting the cancer cell growth. In an embodiment the compound may be selected from formula (1 ), (1A), (1 B) (1C) and (1 D). In an embodiment, the compound comprises a triterpene backbone, two angeloyl groups and sugar moiety. In an embodiment, the compound comprises a triterpene backbone, two acetyl groups with 2 or more carbon and sugar moiety. In an embodiment the compound(s) are selected from Xanifolia (YO, Y1 , Y2, Y, Y7, Y8, Y9, and Y10). In an embodiment the compound(s) are selected from Xanifolia (x), Escin or Aescin. In an embodiment the compound(s) are selected from Compound A to X and A1 to X1 in the application.

In embodiment the method is blocking the cancer cell proliferates by regulating, interacting or affecting the aquaporin. In embodiment, the method is increase the water permeability of the cell membrane by regulating or affecting the aquaporin in order to kill the cell. In an embodiment, the method is affecting the aquaporin permitting extra water into the cell to damage the cancer cell. In an embodiment, the method is affecting the aquaporin permitting Glycerol related solute into the cell to damage the cancer cell. In an embodiment, the method is affecting the aquaporin regulating water into the cell to damage the cancer cell, wherein the method comprise compound selecting from Xanifolia (YO, Y1 , Y2, Y, Y7, Y8, Y9, and Y10).

In embodiment, the method is adjusting the fluid density outside the cell in order to damage the cancer cell by regulating the fluid pass in the cell wherein the aquaporin is overexpressed. In an embodiment this invention makes use of the change of cell membrane permeability to damage the cancer cells. The cell membrane permeability can be changed by the overexpression of some aquaporins.

The potency of Xanifolia(Y) is difference in different cancer cells of ovary, cervix, lung, skin and breast because they have variation of Aquaporin (AQPs 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10) at the membrane.

Amount of Aquarpoin (AQP 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10) at the membrane in various types of ovarian cancer cells such as OVCAR3, SKOV3, TOV21 G and ES2 is different , therefore they show different inhibition activity when treated with Xanifolia (Y). Xanifolia (Y) is a saponin comprises a triterpene, two angeloyl groups and sugar moiety. Xanifolia (Y) inhibits tumor growth ( IC50= 1.5-4.5 ug/ml). Xanifolia (X) which has a similar structure to Y but with only one angeloyl group at C22, has less anticancer activity (IC50=6 ug/ml). Removal of sugars from Y (ACH-Y) but retaining the diangeloyl group retains 40% of the anticancer activity (IC50=9.5 ug/ml).

However, removal of both angeloyl groups from Y (AKOH-Y) completely abolishes its anticancer activity (even at 120 ug/ml). The results indicate that diangeloyl groups in compound Ys are important for anti-tumor activity. The diangeloyl groups pay an important role in interacting with the aquaporin for inhibiting cancer growth.

The hemolytic activities of human red blood cells by Xanifolia-Y (#63Y), Escin and SIGMA saponin standard were compared. Y contains two angeloyl groups, Escin has one angeloyl group and SIGMA saponin standard is a mixture of saponins from Quillaia bark. The results show that #63Y (compound Y) has higher hemolytic activity (IC50=1 ug/ml) than Escin or SIGMA saponin standard (IC50=5 ug/ml). See application PCT/US2006/016158, filed April 27, 2006, Dkt# 804-K-PCT, Figure 6 A

In embodiment, the compounds of this invention interact with cancer cells and increase the static charge of the cells. The static charge increase water flow into the cells. The cancer cells are collaped.

This invention describes a method interacting or regulating the protein on the surface of a cell or altering the functional properties of intracellular membranes or regulating the fluid passage through the cell wall or softening the skin or improving the skin structure, comprising administering to a subject.

This invention describes a method of regulating or affecting the aquaporin, wherein the method comprising the uses of compositions comprising a triterpenoidal saponin. In an embodiment, the saponin has triterpenoid, triterpenoidal or other sapongenin, one or more sugar moieties and two angeloyl groups, or at least two side groups selected from the following groups: angeloyl groups, tigloyl groups or senecioyl groups, wherein the side groups are attached to the sapongenin backbone at carbon 21 and 22. Wherein the sugar moiety comprises at least one or more of 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, the method comprise the use of a composition comprising the structures comprising at least two side groups selected from the following groups: angeloyl, tigloyl or senecioyl groups, wherein the side groups are attached to a triterpenoidal, triterpenoid, triterpenoidal or other sapongenin backbone. These compositions are obtainable from the above-identified plants or synthesis.

This invention provides the uses of composition comprising an effective amount of six novel saponin compounds with the diangeloyl groups (Y1 , Y2, Y, Y8, Y9, Y10) and their structures are as follows. They have anti-cancer and hemotylic effect activies. These six compounds were determined in U.S. Serial 10/906303, International Application No. PCT/US2005/031900. and U.S. Serial 11/131551 , the contents of which are incorporated herein by reference.

The formula, chemical name and common name of these compounds are presented in Table 1.

Table 1. Formula, Chemical Name and Common Name of Six Novel Compounds (Y, Y1 , Y2, Y8, Y9, Y10).

This invention provides a bioactive compound Xanifolia X, oleanene triterpenoidal saponin with a trisaccharide chain attached at C-3 and one angeloyl group acylated at C-22. The saponin is isolated from extract of Xanthoceras sorbifolia. The formula of X is C58H9 2 O 22 and the chemical name is: 3-O-{[/?-D-galactopyranosyl (1→2)]-[«-L-arabinofuranosyl (1→3)]-/?-

D-glucuronopyranoside butyl ester}-21-O-acetyl-22-O-angeloyl-3y?,16α,21#22α,28- pentahydroxyolean-12-ene. The chemical structure of compound X is presented in the following figure.

named as Xanifolia X.

This invention provides a bioactive compound Xanifolia YO and the chemical name is: 3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl(1↠’3)-β-D-glucuronopyranosyl-21- O-angeloyl, 22-O-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12- ene,

This invention provides a bioactive compound Xanifolia Y7 and the chemical name is:

Compound Y 7 3-O-[β-D-galactopyranosyl-(1 →2)]-α-L-arabinofuranosyl-(1 →3)-β-D- -glucuronopyranosyl^i-O-angeloyl^δ-O^-methylbutanoyl-Sβ, 15 σ, 16σ, 21/3, 22α, 28- hexahydroxyolean-12-ene

This invention provides a method of treating a mammal for treating cancers comprising administering to said mammal a therapeutically 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 comprise Xanifolia YO, Y1 , Y2, w , Y7,Y8, Y9, Y10, or a salt, ester, metabolite or

derivative thereof. The compounds of this invention can be isolated from natural sources or synthesized.

See experiments results in Figure 1-18 and see PCT/US05/31900, filed Spetember 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, the contents of which are incorporated herein by reference.

A salt of compound comprise sodium salt, potassium salt or calcium salt. A salt of compounds comprise the following:

A salt of compounds for inhibiting venous insufficiency, particularly hemorrhoids or inhibiting leg swelling, or peripheral edema, antilin p mic, 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, 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; for inhibiting cancer growth.

This invention provides the uses of composition comprising an effective amount of a compound selected from formula (1 ):

named as (1 ) or a salt, ester, metabolite or derivative thereof, wherein R1 and R2 comprises angeloyl group; R3 comprises H or OH; R4 comprises CH 2 OR6, and wherein R6 is H; R5 comprises at least one sugar moiety comprising sugar or its derivatives; R8 may be OH. In an embodiment, R1 and R2 comprise an angeloyl group; R3 comprises H or OH; R4 comprises COOR6 wherein R6 is H. In an embodiment, R1 comprises H; R2 comprises an angeloyl group; R3 comprises H or OH; R4 comprises CH 2 OR6 or COOR6; wherein R6 is an angeloyl group. In another embodiment, R4 comprises CH 2 OR6 or COOR6; at least two of R1 , R2, and R6 comprise an angeloyl group or an acid having five carbons; R3 represents H or OH; and wherein R6 is an angeloyl group, H, acetyl group, tigloyl group, senecioly group, or an acid having two to five carbons. In an embodiment, at least one angeloyl of R1 or R2 is replaced by an acetyl group, tigloyl group, senecioly group, or an acid having two to five carbons; R3 comprises H or OH; R4 comprises CH 2 OR6 or COOR6; and wherein R6 is angeloyl group. In an embodiment, the R4 comprises CH 2 OR6 or COOR6; and wherein R6 is H or acetyl. In an embodiment, at least one of R1 , R2 and R4 comprises a sugar moiety or a side chain comprising sugar or its derivatives, wherein the sugar moiety comprises at least two angeloyl groups, acetyl group, tigloyl group, senecioly group, or an acid having two to five carbons or combination thereof. In a further embodiment, position C23, C24, C25, C26, C29 and C30 independently comprises CH 3 , CH 2 OH, CHO, COOH, alkyls group, acetyl group or their derivatives thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises one or more sugar of, but 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, wherein the sugar moiety comprises two sugars selected

from D-glucose, D-galactose, L-rhamnose, L-arabinose, D-xylose, or their alduronic acids: D-glucuronic acid, D-galacturonic acid, and their derivatives thereof, and the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises three sugars selected from D-glucose, D-galactose, L-rhamnose, L-arabinose, D- xylose, derivatives: alduronic acid, D-glucuronic acid, D-galacturonic acid, and their derivatives thereof, and the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises three sugars selected from D-glucose, D- galactose, L-rhamnose, L-arabinose, D-xylose, and their derivatives thereof, and the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises at least one sugar selected from D-glucose, D-galactose, L-rhamnose, L- arabinose, D-xylose, or alduronic acid: D-glucuronic acid, D-galacturonic acid or derivatives thereof, or the combination thereof. In an embodiment, R5 comprises a sugar moiety comprising glucose, galactose, arabinose or their aldironic acids thereof, or their combination thereof. In an embodiment, R5 comprises a side chain capable of performing the function of the sugar moiety. In an embodiment, the R5 represents H. In a further embodiment, R4 represents H, OH or CH 3 . In a further embodiment, R1 or/and R2 is a functional group capable of performing the function of the angeloyl. R5 comprises a side chain capable of performing the function of the sugar moiety.

A sugar moiety is a side chain (segment of molecule) comprising one or more sugars or their aldironic acids thereof, or derivative thereof. Substitution, deletion and/or addition of any group or groups in the above-described compounds by other group or groups 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.

This invention provides uses of composition comprising effective amounts of compounds selected from formula (1A):

also named as (1A), or a salt, acid, ester, metabolite or derivative thereof, wherein R1 and R2 independently comprise an angeloyl group; R3 comprises H or OH; R4 comprises CH 2 OR6; and wherein

R6 is H; R8 may be OH; R5 comprises at least one sugar moiety or its derivatives. In an embodiment, R1 and R2 independently cor-nrise an angeloyl group; R3 comprises H or OH;

R4 comprises COOR6 wherein R6 is H; R5 comprises at least one sugar moiety or its derivatives. In an embodiment, R1 comprises H; R2 comprises angeloyl group; R3 comprises H or OH; R4 comprises CH 2 OR6 or COOR6; wherein R6 is an angeloyl group; and R5 comprises at least one sugar moiety or its derivatives. In another embodiment, R3 comprises H or OH; R4 comprises CH 2 OR6 or COOR6; and wherein R6 is an angeloyl group, H, acetyl group, tigloyl group, senecioyl group, or an acid having two to five carbons; at least two of R1 , R2, and R6 comprise an angeloyl group or an acid having five carbons; R5 represents at least one sugar moiety or its derivatives. In an embodiment, at least one angeloyl from R1 or R2 is replaced by an acetyl group, tigloyl group, senecioyl group, or an acid having two to five carbons; R3 represents H or OH; R4 comprises CH 2 OR6 or COOR6 wherein R6 is angeloyl group; R5 comprises at least one sugar moiety or its derivatives. In an embodiment, R4 comprises CH 2 OR6 or COOR6; at least one of R1 , R2, and R6 is a sugar moiety comprising at least two angeloyl groups, acetyl group, tigloyl group, senecioyl group, or an acid having two to five carbons or combination thereof. In an embodiment, position 24 of the compound comprises CH 3 or CH 2 OH. In an embodiment, position C23, C24, C25, C26, C29, and C30 of the compound independently comprises CH 3 Or CH 2 OH. In an embodiment, position C23, C24, C25, C26, C29 and C30 of the compound independently comprises CH 3 , CH 2 OH, CHO, COOH, COO-alkyl, COO-aryl, COO- heterocyclic, COO-heteroaryl, CH 2 O-aryl, CH 2 O-heterocyclic, CH 2 O- heteroaryl, alkyls group, acetyl group or derivative thereof. In an embodiment, R5 comprises a sugar moiety and alduronic acid selected from glucose, galactose, arabinose, glucuronic acid, galacturonic acid, or derivative thereof, or the combination thereof. In an embodiment, R5 comprises a 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 or D-xylose, or alduronic acid: D-glucuronic acid, D-galacturonic acid, or derivatives thereof, or the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises two sugars comprising but not limited to, D-glucose, D-galactose, L-rhamnose, L- arabinose or D-xylose, or alduronic acid: D-glucuronic acid, D-galacturonic acid, or derivative thereof, or the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises at least three sugars selected from but not limited to, D-glucose, D-galactose, L-rhamnose, L-arabinose, D-xylose, or alduronic acid: D- glucuronic acid, D-galacturonic acid, or derivative thereof, or the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises at least one sugar of, but is not limited to, glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose or fructose, or alternatively alduronic acid, glucuronic acid, galacturonic acid, or derivative thereof, or the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises three sugars selected from, but not limited to, glucose,

galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose or fructose, or alduronic acid: glucuronic acid and galacturonic acid, or/and derivative thereof, or/and the combination thereof. In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises three sugars selected from, but not limited to, glucose, galactose, rhamnose, arabinose, xylose or fucose, or/and derivative thereof, or/and the combination thereof. In an embodiment, R5 comprises a compound capable of performing the function of the sugar moiety. In a further embodiment, the R5 represents H. In a further embodiment, R4 represents H or OH or CH 3 . In an embodiment, R1 or/and R2 is a functional group capable of performing the function of the angeloyl group. R5 represents a side chain capable of performing the function of the sugar moiety. In an embodiment, R1 and R2 are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl. In an embodiment, R1 and R2 comprise angeloyl, tigloyl, senecioyl, benzoyl or alkenoyl. In an embodiment, R4 represents CH 2 OR6; at least two of R1 , R2 and R6 are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl. In an embodiment, R4 represents CH 2 OR6; at least two of R1 , R2 and R6 are selected from angeloyl, tigloyl, senecioyl, benzoyl or alkenoyl. In an embodiment, R4 represents CH 2 OR6; at least two of R1 , R2 and R6 are selected from angeloyl, benzoyl or alkenoyl. In an embodiment, R1 and R2 are selected from H, angeloyl, acetyl, tigloyl, senecioyl, alkyl, acyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, heterocylic or heteroraryl; R4 represents CH 2 OR6 or COOR6; wherein R6 is selected from H, COCH 3 , angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R4 represents CH 2 OR6, COOR6 or CH 2 COOR6; at least two of R1 , R2 and R6 are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, at least two of R1 , R2 and R4 are comprising angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, at least two of R1 , R2 and R4 comprise angeloyl, acetyl, tigloyl, senecioyl, benzoyl or alkenoyl, or a derivative thereof. In an embodiment, at least two of R1 , R2 and R4 comprise angeloyl, tigloyl, senecioyl, benzoyl or alkenoyl, or a derivative thereof. In an embodiment, at least two of R1 , R2 and R4 comprise a side chian capable of performing the function of the angeloyl group. In an embodiment, at least two of R1 , R2 and R4 comprise a side chian capable of performing the function of benzoyl. In an embodiment, R4 represents CH 2 OR6,

COOR6 or CH 2 COOR6; at least two of R1. R2 and R6 are selected from angeloyl, tigloyl,

senecioyl, benzoyl, alkenoyl, benzoyl or derivatives thereof. In an embodiment, R4 represents CH 2 OR6, COOR6 or CH 2 COOR6; R1 , R2 and/or R6 is/are a sugar moiety comprising groups selected from H, angeloyl, acetyl, tigloyl, senecioly, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R4 represents CH 2 OR6, COOR6 or CH 2 COOR6; R1 , R2 and/or R6 is/are a sugar moiety comprising at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R4 represents CH 2 OR6, COOR6 or CH 2 COOR6; R1 , R2 and/or R6 is/are a sugar moiety comprising at least 2 groups selected from angeloyl, tigloyl, senecioyl, benzoyl, alkenoyl or a derivative thereof. In an embodiment, R4 represents CH 2 OR6, COOR6 or CH 2 COOR6; R1 , R2 and/or R6 is/are a sugar moiety comprising at least 2 groups selected from angeloyl, benzoyl, alkenoyl or a derivative thereof. In an embodiment, a compound selected from formula (1A) comprising at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, or derivative thereof or a group capable of performing the function of angeloyl. In an embodiment, a compound selected from a formula (1A) comprises at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, a compound selected from a formula (1A) comprises a sugar moiety or a side chain of performing function of sugar moiety comprising at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, or a derivative thereof or a group capable of performing the function of angeloyl. In an embodiment, a compound selected from a formula (1A) comprises a sugar moiety or a side chain capable of performing the function of a sugar moiety comprising at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, a compound selected from a formula (1A) wherein R1 and R2 comprises a group selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, acyl, heterocylic or heteroraryl or derivative thereof. R4 is a group 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 or a derivative thereof. A sugar moiety is a segment of a molecule comprising one or more sugars or their aldironic acids thereof, or a derivative thereof. In a further embodiment, the compounds of this invention can be isolated from natural sources or synthesized.

Substitution, deletion and/or addition of any group in the above-described by other groups 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. 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 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.

This invention provides uses of a compound selected from a compound with formula (1 B):

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, aryl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or 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 cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6 or derivative thereof; R4 represents CH 2 OR6, COOR6, wherein R6 is selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or derivative thereof; R3 is H or OH; In an embodiment, R3 comprises alkly, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, acyl, alkanoyl, cycloalkanoyl, alkenoyl, cycloalkenoyl, aryloyl, heteroaryloyl; R5 comprises a 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, R5 comprises hydrogen, alkly, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, acyl, alkanoyl, cycloalkanoyl, alkenoyl, cycloalkenoyl, aryloyl or heteroaryloyl. In an embodiment, R1 comprises a sugar moiety 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, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or a derivative

thereof. In an embodiment, R1 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, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or 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 or heteroraryl , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or 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 or heteroraryl , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein R6 is a sugar moiety which comprises at least one group selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6, wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substitutedalkanoyl, aryl, acyl, heterocylic or heteroraryl , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6, or a derivative thereof. In an embodiment, R4 represents CH 2 OR6, COOR6, wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl, senecioyl, or alkyl. In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein R6 is a sugar moiety which comprises at least two groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, dibenzoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl , acid with carbon 2-6, or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 of formula (1 B), at least two of R1 , R2 and R6 comprise the group selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6, or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 of formula (1 B), 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 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 , cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6, or a derivative thereof. In an 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 CH 3 . In an embodiment, 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. In an embodiment, C23, C24, C25, C26, C29 and C30 of the compound independently comprise alkly, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, acyl, alkanoyl, cycloalkanoyl, alkenoyl, cycloalkenoyl, aryloyl or heteroaryloyl; 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 has two benzoly groups. In an embodiment, R 3 represents H or OH. In an embodiment, R8 may be OH. In an embodiment, R8 comprises alkly, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, acyl, alkanoyl, cycloalkanoyl, alkenoyl, cycloalkenoyl, aryloyl, heteroaryloyl. In embodiment, the R3, R4, R8 or R5 comprise N. In embodiment, the spatially adjacent OH groups derivatized to form a cyclic five or six- membered ring comprising dioxole or cyclic carbonate.

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. A composition comprising an effective amount of the compound selected from the above formula or a salt, ester, metabolite or derivative thereof as a medicament 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.

This invention provides uses of a compound selected from a compound with formula (1C):

also named as (1C), or a salt, ester, metabolite or derivative thereof, wherein R1 and R2 independently comprise an angeloyl group; R3 represents H or OH; R4 comprises CH 2 OR6, wherein R6 is H; R5 comprises a sugar moiety comprising one or more sugars of D-glucose, D-galactose or its derivatives. R7 represents COOH. In an embodiment, R1 and R2 independently comprise an angeloyl group; R3 represents H or OH; R4 comprises COOR6 wherein R6 is H; R5 comprises a sugar moiety comprising one or more sugars of D-glucose, D-galactose or its derivatives. R7 represent COOH. In an embodiment, R1 represents H; R2 comprises angeloyl group; R3 represents H or OH; R4 comprises CH 2 OR6 or COOR6, wherein R6 comprises an angeloyl group or acetyl group. In an embodiment, at least two of R1 , R2, and R6 comprise an angeloyl group or acid having five carbons; R3 represents H or OH; R4 comprises CH 2 OR6 or COOR6, wherein R6 is angeloyl group, H, acetyl group, tigloyl group, senecioyl group, or an acid having two to five carbons. In an embodiment, at least one angeloyl from R1 or R2 is replaced by an acetyl group, tigloyl group, senecioyl group, or an acid having two to five carbons; R3 represents H or OH; R4 represents CH 2 OR6 or COOR6, wherein R6 is angeloyl group. In an embodiment, R4 comprises CH 2 OR6 or COOR6; at least one of R1 , R2, and R6 is a sugar moiety comprising at least two angeloyl groups, acetyl group, tigloyl group, senecioyl group, or an acid having two to five carbons or combination thereof. In an embodiment, position C24 of the compound comprises CH 3 or CH 2 OH. In an embodiment, R7 represents 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, acetyl group or derivatives thereof. In an embodiment, position of C23, C24, C25, C26, C29 and C30 of the compound indepentently comprises CH 3 or CH 2 OH. In an embodiment, position of C23, C24, C25, C26, C29 and C30 of the compound respectively comprise 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, acetyl group or derivative thereof. In an embodiment, R5 comprises a sugar moiety comprising one or more sugars of glucose, galactose, rhamnose, arabinose, xylose, fucose, allose, altrose, gulose, idose, lyxose, mannose, psicose, ribose, sorbose, tagatose, talose, fructose, or alternatively alduronic acid or glucuronic acid or galacturonic acid, or derivative thereof, or the combination thereof. In an embodiment, the R5 represents H. In an embodiment, R4 comprises H, OH, or CH 3 CH 2 OR6, wherein R6 comprises H or an acetyl group. In an embodiment, R1 or/and R2 is a functional group capable of performing the function of the angeloyl. R5 represents a side chain capab lω of performing the function of the sugar moiety.

In an embodiment, R1 and R2 are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivatives thereof. In an embodiment, R1 or/and R2 is a sugar moiety comprising two of groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivatives thereof. In an embodiment, R1 and R2 are selected from H, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl; R4 represents CH 2 OR6 or COOR6, wherein R6 is selected from H, COCH 3 , angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivatives thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 or CH 2 COOR6; at least two of R1 , R2 and R6 are selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivatives thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 or CH 2 COOR6; R1 , R2 and/or R6 is/are a sugar moiety, which comprises at least 2 groups selected from H, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivatives thereof. In an embodiment, a compound selected from formula (1C) comprises at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, or derivatives thereof or a group capable of performing the function of angeloyl. In an embodiment, a compound selected from formula (1C) comprises at least 2 groups selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivatives thereof. In an embodiment, a compound selected from formula (1C) wherein R1 and R2 comprise groups selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. R4 is a compound comprising CH 2 OCOCH 3 , CH 2 COO-alkyl, CH 2 OH, COOH, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof.

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.

This invention provides uses of a compound selected from a compound of formula (1 D):

and also named as (1 D), or a salt, ester, metabolite or derivative thereof, wherein R1 comprise a compound selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof; R2 comprise a compound selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof; R4 comprises CH 2 OR6, COOR6 wherein R6 is selected from hydrogen, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof; R3 is H or OH; R5 comprises a sugar moiety, or D-glucose or D- galactose; R7 represents COOH. In an embodiment, R7 comprises CH 3 , CH 2 OH, CHO, COOH, COOalkyl, COOaryl, COO-heterocyclic, COO-heteroaryl, CH 2 Oaryl, CH 2 O- heterocyclic, CH 2 O- heteroaryl, alkyls group, acetyl group or a derivative thereof. In an embodiment, R1 represents a compound comprising a sugar moiety comprising at least two compounds selected from, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R1 represents a compound comprising a sugar moiety comprising at least one compound selected from, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R2 represents a compound comprising a sugar moiety comprising at least one compound selected from, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R2 represents a compound comprising a sugar moiety or a compound which comprises at least two compounds selected from, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein R6 is a sugar moiety which comprises at least one compound selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein R6 is a sugar moiety which comprises at least two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein R6 is a sugar moiety which

comprises at least two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, or alkyl; In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein R6 is a sugar moiety which comprises at least two compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, dibenzoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof; In an embodiment, R4 comprises CH 2 OR6, COOR6 wherein at least two of R1 , R2 and R6 comprise the compound selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or derivative thereof; In an embodiment, R4 is a compound comprising CH 2 OCOCH3, CH 2 COOalkyl, CH 2 OH, COOH, angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic or heteroraryl or a derivative thereof. In an embodiment, R5 comprises 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 derivative thereof, or the combination thereof. In an embodiment, R5 comprises a compound capable of performing the function of the sugar moiety. In a further embodiment, the R5 comprises a H. In a further embodiment, R4 represents H or OH or CH 3 .

In an embodiment, position 24 of the compound comprise CH 3 or CH 2 OH, In a further embodiment, positions 23, 24, 25, 26, 29, 30 of the compound independently comprise CH 3 ,

CH 2 OH, CHO, COOH, COOalkyl, COOaryl, COO-heterocyclic, COO-heteroaryl, CH 2 Oaryl,

CH 2 O- heterocyclic, CH 2 O- heteroaryl, alkyls group, acetyl group or a derivative thereof. In an embodiment, R5 comprises a sugar moiety comprising L-glucose, D-galactose, L- rhamnose, or/and L-arabinose. In an embodiment, R1 and R2 independently comprise an angeloyl group; In a embodiment, R1 is a sugar moiety or rhamnose which comprise two angeloyl groups.

In an embodiment, R3 represents H or OH; In an embodiment, the compounds can be isolated from natural sources or synthesized.

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 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, dysmenorrhea!, 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 compounds in Figure 1 to 3, 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.

This invention provides a method of inhibiting tumor cell growth comprising administering to a subject, in need thereof, an appropriate amount of triterpenoidal saponins comprising two or more angeloyl groups or comprising the structure of Figures 1-3, 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 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.

In an embodiment of the above, the uses of compositions for medicament comprising any one of triterpenoid saponins with the following formula:

3-O-{[/?-D-galactopyranosyl (1 →2)]-[α-L-arabinofuranosyl (1 →3)]-/?-D-glucuronopyranoside butyl ester}-21-O-acetyl-22-O-angeloyl-3y?,16«,21y?,22«,28-penta hydroxyolean-12-ene. 3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl(1↠’3)-β-D-glucuronopyranosyl- 21 ,22-O-diangeloyl-3β, 15α, 16α, 21β, 22α, 28-hexahydroxyolean-12-ene,

3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl( 1→3)-β-D-glucuronopyranosyl -21-

O-(3,4-diangeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β, 16α, 21 β, 22α, 28- pentahydroxyolean-12-ene,

3-O-[β-D-glucopyranosyl-(1→2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl- 21 ,22-O-diangeloyl-3β, 15α, 16α, 21β, 22α, 24β, 28-heptahydroxyolean-12-ene,

3-O-[/?-glucopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-0- diangeloyl-3/?, 16«, 21/?, 22«, 24/?, 28-hexahydroxyolean-12-ene,

3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21-O-(3,4- diangeloyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12- ene,

3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-0- diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene,

3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl( 1→3)-β-D-glucuronopyranosy 1-21-

O-angeloyl, 22-O-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12- ene,

3-O-[β-D-galactopyranosyl-(1→2)]-α-L-arabinofuranosyl -(1 →3)-β-D-

-glucuronopyranosyl^i-O-angeloyl^δ-O^-methylbutanoyl-Sβ , 15 a, 16σ, 21/3, 22α, 28- hexahydroxyolean-12-ene

This invention provides a composition comprising the compounds as described above effective in inhibiting cancer growth. The cancer includes but is not limited to bladder cancer, bone, cancer, skin cancer and ovarian cancer.

This invention also provides a composition for inhibiting venous insufficiency, particularly hemorrhoids or inhibition of leg swelling, or inhibiting cancer growth comprising any of compounds selected from the following compounds:

A) 3-O-[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,

B) 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl (1→3)-β-D-glucuronopyranosyl- 21-O-(3, 4-diangeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene

C) 3-O-[β-D-glucopyranosyl-(1→2)]-α-L-arabinofuranosyl (1 →3)-(-D-glucuronopyranosyl-21 , 22-O-diangeloyl-3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12-ene

D) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22- O-diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene

E) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1^3)-/?-glucuronopyranosyl-21-O- (3,4-diangeloyl)-«-rhamnopyranosyl-28-O-acetyl-3$ 16«, 2 * \β, 22«, 28-pentahydroxyolean- 12-ene

F) 3-O-[/?-galactopyranosyl (1 →2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22- O-diangeloyl-3# 16«, 2^β, 22«, 28-pentahydroxyolean-12-ene

G) 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl (1→3)-β-D-glucuronopyranosyl - 21 , 22-O-dibenzoyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene,

H) 3-O-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl-

21-O-(3,4- dibenzoyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene

I) 3-O-[β-D-glucopyranosyl-(1→2)]-α-L-arabinofuranosyl (1 →3)-(-D-glucuronopyranosyl -21 ,

22-0- dibenzoyl -3β, 15«, 16«, 21y?, 22«, 24jS, 28-heptahydroxyolean-12-ene

J) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-

O- dibenzoyl -3jβ, 16«, 21y?, 22«, 28-pentahydroxyolean-12-ene K) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21-O-

(3,4-dibenzoyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 2 * \β, 22«, 28-pentahydroxyolean-

12-ene

L) 3-O-[/?-galactopyranosyl (1 →2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-

O- dibenzoyl -3β, 16«, 2^β, 22«, 28-pentahydroxyolean-12-ene M) 3-O-[β-D-galactopyranosyl (1 →2)]-β-D-xyopyranosyl (1 →3)-β-D-glucuronopyranosyl -21 ,

22-O-dibenzoyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene,

N) 3-O-[β-D-galactopyranosyl(1→2)]- β-D-xyopyranosyl (1→3)-β-D-glucuronopyranosyl-

21-O-(3,4- dibenzoyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene O) 3-O-[β-D-glucopyranosyl-(1→2)]-β-D-xyopyranosyl (1→3)-(-D-glucuronopyranosyl -21 ,

22-0- dibenzoyl -3β, 15«, 16«, 2 ^ \β, 22«, 24y?, 28-heptahydroxyolean-12-ene

P) 3-O-[/?-D-galactopyranosyl (1→2)]-β-D-xyopyranosyl (1→3)-/?-D-glucuronopyranosyl -21 ,

22-0- dibenzoyl -3β, 16«, 2 ^ \β, 22«, 28-pentahydroxyolean-12-ene

Q) 3-O-[/?-galactopyranosyl (1 ^2)]- β- xyopyranosyl (1→3)-/?-glucuronopyranosyl-21-O- (3,4- dibenzoyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21$ 22«, 28-pentahydroxyolean-

12-ene

R) 3-O-[/?-galactopyranosyl (1→2)]-β- xyopyranosyl (1→3)-/?-glucuronopyranosyl-21 , 22-0- dibenzoyl -3β, 16«, 2^β, 22«, 28-pentahydroxyolean-12-ene

S) 3-O-[β-D-galactopyranosyl (1→2)] - β- D-xyopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 , 22-O-diangeloyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene,

T) 3-O-[β-D-galactopyranosyl (1→2)] - β- D-xyopyranosyl (1→3)-β-D-glucuronopyranosyl- 21-O-(3,4-diangeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β, 16α, 21 β, 22α, 28- pentahydroxyolean-12-ene

U) 3-O-[β-D-glucopyranosyl-(1→2)] - β- D-xyopyranosyl (1→3)-(-D-glucuronopyranosyl-21 , 22-O-diangeloyl-3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12-ene

V) 3-O-[/?-galactopyranosyl (1 →2)] - β- D-xyopyranosyl (1→3)-/?-glucuronopyranosyl-21 , 22- O-diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene

W) 3-O-[/?-galactopyranosyl (1→2)] - β- D-xyopyranosyl (1→3)-/?-glucuronopyranosyl-21-O- (3, 4-diangeloyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean- 12-ene

X) 3-O-[/?-D-galactopyranosyl (1→2)] - β- D-xyopyranosyl (1→3)-/?-D-glucuronopyranosyl- 21 , 22-O-diangeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene

This invention provides a composition for inhibiting cancer growth comprising any of the compounds selected from the following:

A1 ) 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl(1→3)-β-D-glucuronopyranosy l- 21-O-angeloyl,22-O-benzoyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene, B1 ) 3-O-[β-D-galactopyranosyl (1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D- glucuronopyranosyl-21-O-(3-angeloyl, 4-benzoyl)-α-L-rhamnophyranosyl-22-O-acetyl- 3β,16α, 21 β, 22α, 28-pentahydroxyolean-12-ene

C1 ) 3-O-[β-D-glucopyranosyl-(1→2)]-α-L-arabinofuranosyl (1→3)-(-D-glucuronopyranosyl- 21-O-angeloyl,22-O-benzoyl-3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12-ene D1 ) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1 →3)-/?-glucuronopyranosyl-21-O- angeloyl, 22-benzoyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene E1 ) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21-O- F) (3-angeloyl, 4-benzoyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28- pentahydroxyolean-12-ene

F1 ) 3-O-[/?-galactopyranosyl (1 →2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21-O- angeloyl, 22-O-benzoyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene G1) 3-O-[β-D-galactopyranosyl (1→2)]-α-L-arabinofuranosyl (1→3)-β-D- glucuronopyranosyl-21 -O-benzoyl, 22-O-angeloyl-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene,

H1 ) 3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl(1↠’3)-β-D-glucuronopyranosyl- 21-O-(3- benzoyl, 4-angeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene

11 ) 3-O-[β-D-glucopyranosyl-(1 →2)]-α-L-arabinofuranosyl (1 →3)-(-D-glucuronopyranosyl-21 -O-benzoyl, 22-O-angeloyl-3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12-ene J1 ) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 -O- benzoyl, 22-O-angeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene K1 ) 3-O-[/?-galactopyranosyl (1→2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21-O- (3- benzoyl, 4-angeloyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28- pentahydroxyolean-12-ene

L1 ) 3-O-[/?-galactopyranosyl (1 →2)]-«-arabinofuranosyl (1→3)-/?-glucuronopyranosyl-21 -O- benzoyl, 22-O-angeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene M1 ) 3-O-[β-D-galactopyranosyl (1→2)]-β-D-xyopyranosyl (1→3)-β-D-glucuronopyranosyl-21 -O-angeloyl, 22-O-benzoyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene, N1 ) 3-O-[β-D-galactopyranosyl(1→2)]- β-D-xyopyranosyl (1→3)-β-D-glucuronopyranosyl- 21-O-(3-angeloyl, 4- dibenzoyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β,16α, 21 β, 22α, 28- pentahydroxyolean-12-ene 01) 3-O-[β-D-glucopyranosyl-(1→2)]-β-D-xyopyranosyl (1 →3)-(-D-glucuronopyranosyl-21 - 0-21 -O-angeloyl, 22-O-benzoyl -3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12- ene,

P1 ) 3-O-[/?-D-galactopyranosyl (1→2)]- β- D-xyopyranosyl (1→3)-/?- D-glucuronopyranosyl- 2121 -O-angeloyl, 22-O-benzoyl -3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene Q1) 3-O-[/?-galactopyranosyl (1→2)]- β- xyopyranosyl (1→3)-/?-glucuronopyranosyl-21-O-(3- angeloyl, 4- dibenzoyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28- pentahydroxyolean-12-ene,

R1 ) 3-O-[/?-galactopyranosyl (1→2)]-β- xyopyranosyl (1→3)-/?-glucuronopyranosyl~angeloyl, 22-O-benzoyl -3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene, S1 ) 3-O-[β-D-galactopyranosyl (1^2)] - β- D-xyopyranosyl (1 →3)-β-D-glucuronopyranosyl- 21 -O-benzoyl, 22-O-angeloyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene, T1 ) 3-O-[β-D-galactopyranosyl (1^2)] - β- D-xyopyranosyl (1→3)-β-D-glucuronopyranosyl- 21-O-(3-benzoyl, 4-angeloyl)-α-L-rhamnophyranosyl-22-O-acetyl-3β, 16α, 21 β, 22α, 28- pentahydroxyolean-12-ene, U1 ) 3-O-[β-D-glucopyranosyl-(1→2)] - β- D-xyopyranosyl (1→3)-(-D-glucuronopyranosyl-21 -O-benzoyl, 22-O-angeloyl-3/?, 15«, 16«, 21/?, 22«, 24/?, 28-heptahydroxyolean-12-ene V1 ) 3-O-[/?-galactopyranosyl (1^2)] - β- D-xyopyranosyl (1→3)-/?-glucuronopyranosyl-21 - O-benzoyl, 22-O-angeloyl-3/?, 16«, 21/?, 22«, 28-pentahydroxyolean-12-ene W1 ) 3-O-[/?-galactopyranosyl (1 ^2)] - β- D-xyopyranosyl (1→3)-/?-glucuronopyranosyl-21- O- (3-benzoyl, 4-angeloyl)-«-rhamnopyranosyl-28-O-acetyl-3/?, 16«, 21/?, 22«, 28- pentahydroxyolean-12-ene

X1 ) 3-O-[/?-D-galactopyranosyl (1→2)] - β- D-xyopyranosyl (1→3)-/?-D-glucuronopyranosyl- 21 -O-benzoyl, 22-O-angeloyl-3# 16«, 2λβ, 22a, 28-pentahydroxyolean-12-ene.

This invention provides a composition for inhibiting cancer growth comprising any of the compounds selected from the following:

1 ) 3-O-[β-D-galactopyranosyl-(1 →2)]-α-L-arabinofuranosyl-(1 →3)-β-D- -glucuronopyranosyl-21-O-angeloyl-22-O-(angeloyl-2-methylbut anoyl) -3β, 15 a, 16α, 21/3, 22a, 28-hexahydroxyolean-12-ene

2) 3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl(1↠’3)-β-D-glucuronopyranosyl- 21-O-(2-methylpropanoyl), 22-O-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28- hexahydroxyolean-12-ene,

3) 3-O-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl- 21-O-angeloyl, 22-O-benzoyl-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene,

4) 3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl(1↠’3)-β-D-glucuronopyranosyl- 21-O-(2-methylpropanoyl)-O-benzoyl, 22-O-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α,

28-hexahydroxyolean-12-ene,

5) 3-O-[β-D-galactopyranosyl(1 →2)]-α-L-arabinofuranosyl(1 →3)-β-D-glucuronopyranosyl- 21-O-(2-methylpropanoyl)-O-angeloyl, 22-O-(2- methylbutanoyl) -3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12-ene,

Triterpenoid saponins with the characteristic structures mentioned above in this invention can be used to inhibit venous insufficiency, particularly hemorrhoids or inhibit leg swelling, Triterpenoid saponins with the characteristic structures mentioned above in this invention can be used to reduce or inhibit cancer growth. 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. Triterpenoid saponins with the characteristic structures mentioned above in this invention can be used to affect cell membrane structure and adhesion process. In an embodiment, it provides a method of binding with and adhesion proteins to blocks the migration, metastasis of cancer cells, and growth of cancers.

In an embodiment, the compound is a triterpenoidal saponin or sapongenin, wherein the triterpenoidal saponin comprises at least any one or two of an angeloyl group, tigloyl group, or senecioyl group, or their combinations thereof at carbon 21 and/or 22, or 28, directly attached to the sapogenin or attached to a sugar moiety can be used to to treat varicose vein disease, inhibit venous insufficiency, particularly hemorrhoids or inhibit leg swelling, reduce or inhibit cancer growth. In an embodiment, the compound is a five ring triterpene

saponin comprising at least two angeloyl groups, tigloyl group, or senecioyl group, or their combinations thereof and a sugar moiety. The angeloyl groups are attached to a side chain at the end of the five rings and a sugar moiety is attached to a side chain of the ring at the other end of the five rings. In an embodiment, the compound comprises at least two angeloyl groups, a tigloyl group, or a senecioyl group, or combinations thereof and a sugar moiety. The angeloyl groups and the sugar moiety are attached to the side chains of the backbone of the compound respectively. In an embodiment, the angeloyl can be replaced by a functional group which functions as an angeloyl group. In an embodiment, a sugar moiety or chain is at C3 or other positions, comprising 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 preferably D- glucose, D-galactose, L-rhamnose, L-arabinose, alduronic acids of D- glucuronic acid or D-galacturonic acid, or their combinations thereof, or their derivatives thereof. In a further embodiment, CH 3 or CH 2 OH or COOH or acetyl group may attach at C 23-30 independently. The activities of a saponin compound for regulating or inhibiting tumor cell growth are based on or attributed to its structure that has the functional group(s) such as angeloyl group, tigloyl group, senecioyl group or acetyl group, or their combinations thereof.

Both compound Y1 and Compound Y2 have two angeloyl groups and therefore, strongly inhibited the growth of cancer cells (See Figure 4).

The compounds Y, Y1 , Y2, Y8, Y9 and Y10 which all have two angeloyl groups and, therefore, inhibited the growth of ovarian cancer cells (See Figure 5).

The compound (X) and Escin with single angeloyl groups showed weaker anticancer activity and hemolytic activity compared with the compounds with two angeloyl groups (See Figure 5, 6 and 7).

The compound without angeloyl groups has no anticancer and hemolytic activies (See

Figure 4, 5, 6 and 7).

The compound with two angeloyl groups have stronger potency than the one with one angeloyl for 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 and peripheral vascular disorders.

This invention provides a composition comprising the compounds with the structure of:

wherein R1 and R2 comprise angeloyl groups, tigloyl groups, senecioyl groups or acetyl group or their combinations, preferable wherein the R1 and R2 comprise angeloyl groups. In an embodiment, R1 and R2 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative thereof.

wherein R1 and R2 comprise angeloyl groups, tigloyl groups, senecioyl groups or acetyl group or their combinations, preferable wherein the R1 and R2 comprise angeloyl groups.

In an embodiment, R1 and R2 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative thereof.

In an embodiment, the compound further comprises a sugar moiety.

In a further embodiment, the sugar moiety comprises glucose, galactose or arabinose or combination thereof.

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 R1 or R 2 may be attached in other position of the structure.

wherein 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 embodiment, at least two of R1 , R2 and R3 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl,

alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 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, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative thereof.

In an embodiment, the compound comprises a sugar moiety. In an embodiment, the sugar moiety is attached at one end of structure (d), opposite to R1 , R2 and R3. 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. In an embodiment, the R1 , R 2 and R3 may be attached in other position of the structure.

In an embodiment, the compound is triterpenoid 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, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 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, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 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.

In an embodiment, a triterpene comprise the following structure has anti-cancer or inhibiting virus activities.

wherein 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 embodiment, at least two of R1 , R2 and R3 comprise compounds selected from angeloyl, acetyl, tigloyl, senecioyl, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 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, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative thereof.

In an embodiment, R5 comprises a sugar moiety, wherein the sugar moiety comprises of glucose, galactose or arabinose or alduronic acid 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 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 R1 , R 2 and R3 may be attached in other position of the structure.

In an embodiment, the compound is triterpenoid 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, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 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, alkyl, benzoyl, dibenzoyl, alkanoyl, alkenoyl, benzoyl alkyl substituted alkanoyl, aryl, acyl, heterocylic, heteroraryl, cycloalkyl, cycloalkanoyl, cycloalkenoyl, aryloyl, heteroaryloyl, acid with carbon 2-6 or derivative 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 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.

In a further embodiment, a compound or sapongenin comprises the structure (d) or (e) has anti-cancer or inhibiting virus activities.

A composition for 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 alternative carbon in cis position. In an embodiment, the side chains are at alternative carbon in trans position.

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.

This invention provides a composition regulating the protein on the surface of the cell or alters the functional properties of intracellular membranes. The compounds and compositions provided in this invention can regulate the water passing through the cell wall to soften the skin or improve the skin structure.

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; and for treating impotence or premature ejaculation or diabetes (See PCT/US05/31900, filed Spetember 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. 11/131551 , filed May 17, 2005, the contents of which are incorporated herein by reference).

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, antipleuritic, antiraucedo, antirhinitic, antitonsilitic, antiulcer, antivaricose, antivertiginous, cancerostatic, corticosterogenic, diuretic, fungicide, hemolytic, hyaluronidase inhibitor, lymphagogue, natriuretic, pesticide, pituitary stimulant, thymolytic, vasoprotective, and venotonic treatment.

A composition comprising an effective amount of the compound of any one of YO, Y1 , Y2, Y(Y3),Y7, Y8, Y9, Y10, or a salt, ester, metabolite or derivative thereof as a medicament 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.

This composition can be administered orally or in a particular embodiment, it can be administered through intraperitoneal (I.P.) > intravenous (I.V.) injection or intravenous drip.

In an embodiment, the medicine can be administered with glucose solution or NaCI solution. The administration of the medicine can be as intravenous injection or intravenous drip.

Example 1 : Intravenous drip: 0.05-0.2mg/kg medicine dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution.

Example 2: Intravenous injection: 0.05-0.2mg/kg/day medicine dissolved in 10-2OmI of 10% glucose solution or of 0.9% NaCI solution. Course of treatment: 7-10 days.

Example 3: Intravenous drip: 0.1-0.2mg/kg/day medicine dissolved in 250ml of 10% glucose solution or in 250ml of 0.9% NaCI solution. Course of treatment: 7-10 days.

Example 4: Intravenous injection: 0.1-0.2mg/kg/day medicine dissolved in 10-2OmI of 10% glucose solution or of 0.9% NaCI solution. Course of treatment: 7-10 days. Example 5: Intraperitoneal (I. P.): 2.5mg/kg/day medicine dissolved in 10% glucose solution or of 0.9% NaCI solution. Course of treatment: 7-10 days.

The composition can be administered orally wherein the dosage of mammal is 1-10mg/Kg.

The composition can be administered orally wherein the dosage is 10-30mg/Kg.

The composition can be administered orally wherein the dosage is 30-60mg/Kg. The composition can be administered orally wherein the dosage is 60-90mg/Kg.

The composition can be administered intravenous injection or intravenous drip wherein the dosage of mammal is 0.01- 0.1 mg/Kg.

The composition can be administered intravenous injection or intravenous drip wherein the dosage is 0.1-0.2mg/Kg. The composition can be administered intravenous injection or intravenous drip wherein the dosage is 0.2 - 0.4mg/Kg,

The composition can be administered intravenous injection or intravenous drip wherein the dosage is 0.4 - 0.6 mg/Kg.

The composition can be administered intraperitoneal (I. P.) wherein the dosage of mammal is 1-3mg/Kg.

The composition can be administered intraperitoneal (I. P.) wherein the dosage is 3-5mg/Kg.

The composition can be administered intraperitoneal (I. P.) wherein the dosage is 4-6mg/Kg.

The composition can be administered intraperitoneal (I. P.) wherein the dosage is 6-

10 mg/Kg.

This invention provides a method of treating a mammal for treating cancers comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising a composition comprises the molecular formula or compound in this invention. 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. The compounds comprise Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, or a salt, ester, metabolite or derivative thereof. See experiments results in Figure 4-6, Figure 11-23 and Figure 37-38.

This invention describes a method interacting or regulating the protein on the surface of a cell or altering the functional properties of intracellular membranes or regulating the fluid passage through the cell wall to kill the cancer cells. The method comprising administering contacting an effective amount of compound selected from formula (1 ), (1A), (1B), (1C), (1 D) preferable XanifoliaYO, Y, Y1 , Y2, Y7, Y8, Y9, Y10. In an embodiment the compound select from Xanifolia YO, Y, Y1 , Y2, Y7, Y8, Y9, and Y10 interact with the protein in the membrane and open up the channel for water or solute particle. The cell takes in water or solute particle and bursts.

In an embodiment the components of the compound select from XanifoliaYO, Y, Y1 , Y2, Y7,

Y8, Y9, and Y10 combine with the protein in the membrane and open up the channel for water or solute particle. The cell takes in water or solute particle and bursts. One or more aquaporin AQPs 1 , 2, 3, 4, 5, 6, 7, 8, 9, and 10 in the cancer cell membrane is overexpressed. So as providing more chance react with Xanifolia compound. Water or ion particle pass through the cell membrane the cancer cells

The compound select from XanifoliaYO, Y, Y1 , Y2, Y7, Y8, Y9, and Y10 dilute the solution outside the cancer to make more water pass in the cell. The overexpress of Aquaporin of AQPs 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 provide more channel for water or ion particle which causes cancer cell die.

This invention provides a method of inhibiting cancer growth by destroys the cancer cell wherein aquaporin is overexpressed; 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. In an embodiment, the cancer is ovarian cancer.

This invention provides a method of treating a mammal for treating cancers comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising a composition comprises the molecular formula or compound in this invention. 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. The compounds comprise Xanifolia YO, Y1 , Y2, Y, Y7, Y8, Y9, Y10, or a salt, ester, metabolite or derivative thereof. 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 carbocyclic aromatic compound such as benzene, a 6-14 membered carbocyclic aromatic ring system comprising 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, wherein 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, and can be represented as C 6 H 5 CO-R, obtained from benzoic acid by the removal of the carboxyl.

Heterocyclic compound - a compound containing a non-aromatic 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, represented by 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 (e.g. tigloyl) and hexenoyl (e.g. 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 refers 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, lsobutyryl is Synonym of 2-Methylpropanoyl Y and Y3 represent the same compound.

To investigate the anti-tumor activity of compounds in this application, we employed cancer cell lines derived from different human organs and tested the effect on growth activity. In these preliminary studies, we found that the plant extract inhibits the growth of certain cell lines. We studied 10-15 cell-lines (derived from different human organs) with a MTT cell- growth assay, and found that OVCAR3 cells (from ovary) to be the most sensitive (with IC50 = 14.5 ug/ml). Int'l App'l No. PCT/US2004/033359 and U.S. Serial No. 10/906,303.

The active compound was then purified and named Xanifolia-Y. Its chemical structure was determined by 2D NMR and MS analysis. Xanifolia-Y is a novel triterpenoid saponin with a diangeloyl group attached at one end and carbohydrates or sugar moieties at another end of the triterpene structure. In an embodiment, the diangeloyl attached at C21 , C22 positions and carbohydrates or sugar moieties at C3 position of a triterpene structure. The diangeloyl group is important for its activity. The purified compound has been tested with 60 cancer cell lines. Test results show inhibition towards most cell lines tested with GI50 values ranging from 0.1 - 1 uM.

OVCAR3 cells, cancer cells derived from ovary, are the most sensitive to Xanifolia-Y among cell lines tested in our early studies. We subsequently tested 10 additional human ovarian cancer cell lines and found all of them to be susceptible to inhibition by Xanifolia-Y with IC50 values ranging from 2-12 uM. See experiment 11.

In vivo studies employing human ovarian carcinoma xenografts in nude mice were performed. The human ovarian cancer cells (ES2) were inoculated into the peritoneal cavity of nude mice and subsequently received Xanifolia-Y. (Experiment 7, 8, 9). The tumor

bearing mice received the drug (by i.p. route) for 10 days starting from either day 1 , day 4, or day 10 after inoculations. The results show that the median survival time for tumor bearing mice without drug-treatment is approximately 20-24 days. However, there was no death for tumor bearing mice with drug-treatment starting on day 1 after tumor inoculation. The median survival time for tumor bearing mice with drug-treatment starting on day 4 after tumor inoculation is no death in 50 days; and tumor bearing mice with drug-treatment started on day 10 after tumor inoculation is half of the mice survive in 50 days. These results indicate that the compounds of this invention are capable of increasing the survival rate of mammal.

The median survival time for tumor bearing mice with drug-treatment starting on day 4 after tumor inoculation is 58 days (extension of life span of 141 %); and tumor bearing mice with drug-treatment started on day 10 after tumor inoculation is 31 days (extension of life span of 29%).. These results indicate that Xanifolia-Y is capable of extending the life span of mammal bearing tumors. It is useful in treating ovarian cancer in humans.

These results indicate that Xanifolia-Y is capable of extending the life span of mammal bearing tumors. It is useful in treating ovarian cancer in humans.

Among gynecological malignancies, ovarian cancer has the highest rate of mortality in women in the United States with an estimated 22,220 new cases in 2005 and over 16,000 deaths (NIH web info). The disease is often missed in diagnosis in the early stage due to asymptomatic and the lack of reliable diagnostic marker. As a result, most of the ovarian cancer patients being diagnosed are already at advanced stages. The standard treatment of ovarian cancer is a combination of a platinum analogue with paclitaxel (McGuire et al., 1996; Ozols et al., 2003). Improved patients survival time was observed in patients with intraperitoneal administration of these agents (Armstrong et al., 2006). The peritoneal cavity is the principal site of disease in ovarian cancer. The improved efficacy of these agents could be due to a more direct interaction with cancer cells. However, the increase of median survival from 49.7 to 65.6 months is still far from satisfactory.

As mentioned above, our in vivo animal experiments mimicking the human situation showed that Xanifolia-Y is effective in prolonging mammal life span. Mice were inoculated with human ovarian carcinoma (ES2) in the peritoneal cavity. Starting from the mid-way (time to mortality) point of tumor progression (considered as a late stage of disease in human), drug was then administered into the peritoneal cavity. It was found that Xanifolia-Y treatment is beneficial to tumor bearing mice by prolonging their life span. Depending on the stage of the disease progression, the sooner the start of the drug-treatment, the better the results are.

Based on our results, it can prolonged the life-span of tumor bearing mice after Xanifolia-Y- treatment is due to blockage of the migration or metastasis of inoculated cancer cells into the mesothelium lining in the peritoneal cavity. In vitro studies show that Xanifolia-Y inhibits cell adhesion to culture flasks (See Experiment 13). It is known that adhesive molecules play an important role in the migration and metastasis of ovarian cancer (Skubitz, 2002, Schaller, 1996; Zetter, 1993). A major route for the spread of ovarian cancer is by the attachment of tumor cells to the mesothelium lining in the peritoneal cavity (Gardner et al., 1995). Xanifolia-Y blocks the function of these adhesive molecules on cells. In an embodiment, Xanifolia-Y blocks the function of these adhesive molecules on carcinoma cells. In an embodiment, Xanifolia-Y blocks the function of these adhesive molecules on ovarian carcinoma cells. In an embodiment, Xanifolia-Y blocks the function of these adhesive molecules on the mesothelial cells. In an embodiment, Xanifolia-Y binds to the adhesive proteins (by masking) on the membrane and inhibits the interaction of adhesion proteins with their receptors. In an embodiment, Xanifolia-Y action on membrane affects adhesion proteins' function in membrane. The lost of adhesion activity of cancer cells is result from direct or indirect action of Xanifolia-Y on membrane proteins.

Most of the adhesion proteins are glycoproteins. The carbohydrate moiety in adhesion proteins interact with carbohydrates from other molecules, such as saponin or Xanifolia-Y. Xanifolia-Y has a trisaccharide at the C3 position and it was found that a loss of carbohydrates reduces its activity (Fig. 4D and 5C). Our EM studies show that Xanifolia-Y affects membrane structure and makes holes. Damage to the membrane structure could alter adhesion protein's conformation and interfere with their binding with other molecules or even cause them to lose their anchorage on membrane.

Our studies of Xanifolia-Y indicate it can be used in cancer therapy, especially as a benefit to patients with late stage ovarian cancer. They indicate that our determined saponins and formulas are useful in cancer therapy by demonstrating its inhibition of tumor growth in mammal systems.

To study the effect of Xanifolia-Y on membrane structure, the morphology of cell membrane treated with Xanifolia-Y was examined with EM. In this experiment, K562 cells were treated with 5 uM of Xanifolia-Y for 60 min. Solvent DMSO and AKOH-Y (a derivative of Xanifolia-Y without the angeloyl group and it has no activity) served as controls. Cells were negative stained with 1 % UAc and subsequently examined with EM. Figure 25 show that patches of pits were found in the membrane of Xanifolia-Y treated cells (Fig. 25B) but not in cells treated with the DMSO (Fig. 25A) or AKOH-Y (Fig. 25C) controls. These pits have the size from 8OA to 500A (in diameter). The pits represent holes formed in the membrane. The pits are arranged in a characteristic pattern with smaller pits (8OA in diameter) located in the

periphery and the bigger ones (500A in diameter) in the center. The bigger holes are resulted from fusing of the smaller holes (Fig. 25D). Membrane image of cells treated with A: DMSO solvent control, 60 min (magnification: X60,000); B: Xanifolia-Y 5 uM, 60 min. (X60000); C: AKOH-Y, 20 uM, 60 min. (X60000); D: Xanifolia-Y 5 uM, 60 min. (X20000). This experiments results show that the Xanifolia-Y alters the membrane of cell. In an embodiment, it damages the membrane of cancer cell.

Xanifolia-Y can be used for inhibiting cancers cell growth or treating cancers 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 cancer is preferably ovarian cancer. Among the different cell lines tested in our studies, carcinoma cells derived from ovary proved to be the most sensitive, a finding which is substantiated with more human ovarian cancer cell lines. The results of animal studies with human tumor xenograft in mice show that it can extend the life span of mice bearing tumors. The compounds of this application can extend the life span of a subject or mammal that bearing human cancer.

Cancer drugs that target on membrane or membrane constituents are not explored. Xanifolia-Y is a new drug. It has effects on cell membrane, a target that differs from current anticancer drugs.

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 method of inhibiting the growth, migration, metastasis of cancer by altering the characteristic of membrane of cancer cell, wherein the characteristic comprise adhesion protein; 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, 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 invention provides a composition 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.

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 15)

In our animal studies, it was shown that Xanifolia-Y extended the life span of tumor bearing mice. (See Experiments 7, 8, 9, and 10). 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 is blocking of the protein accessibility due to direct binding to Xanifolia-Y, In an embodiment, the interaction of Xanifolia-Y with membrane alter indirectly the adhesion protein's binding site(s).

We have shown that Xanifolia-Y are cytotoxic to tumor cells, in an embodiment it kills ovarian cancer cells. Our inventions show that Xanifolia-Y inhibits cancer cell growth and prolongs life-span of tumor bearing mice. Our studies also indicate that the sooner the drug- treatment, the longer the life-span of the tumor bearing animals is extended. Xanifolia-Y also has an effect in blocking or inhibiting migration or metastasis. The delay of Xanifolia-Y- treatment allows more chances for cancer cells to metastasize to the mesothelium lining in the peritoneal cavity which resulted in more tumor growth and shorter life span. Adhesive molecules play an important role in cell migration and metastasis. It was shown in our studies that Xanifolia-Y inhibits cell attachment to culture flasks. Xanifolia-Y interferes with the function of the adhesive molecules. In embodiment Xanifolia-Y blocks the function of the adhesive molecules. In an embodiment, Xanifolia-Y binds directly to adhesive proteins. It is masking the adhesive proteins. In an embodiment, Xanifolia-Y indirectly alters membrane structure that cause changes in protein conformation, or locations and result in loss of adhesion process.

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 are disclosed in PCT/US2005/031900, U.S. Serial No. 1 1/289,142, U.S. Serial 10/906,303, and U.S. Serial No. 11/131 ,551 , the contents of which are incorporated herein by reference.

Experiment 1 : Determination of the hemolytic activities of compound Y from Xanthoceras sorbitol ia Methods:

1 ) Human whole blood was obtained from the Houston Gulf Coast Blood Center. 2) Red blood cells were isolated by the following method: Human blood (in EDTA) was diluted 1 :1 with PBS, underlay with 4 ml_ of Histopaque-1077 (SIGMA) and was centrifuged at 40Og for 30 min. 3) Red blood cells (RBC) were collected and washed three times with PBS. 4) 10% suspensions of RBC were prepared with PBS before use. 5. 50 μl_ of RBC suspension was added to 2 ml_ of saponins with different concentration. 5) The suspension was mixed by vortexing then left to sit at room temperature for 60 minutes. 6) The suspension was centrifuged at 3000 rpm for 5 min. Absorbance of the supernatant was measured at 540 nm.

Results:

In this experiment, hemolytic activities of human red blood cells by Xanifolia-Y (#63Y), Escin and SIGMA saponin standard were compared. Y contains two angeloyl groups, Escin has one angeloyl group and SIGMA saponin standard is a mixture of saponins from Quillaia bark. The results show that #63Y (compound Y) has higher hemolytic activity (IC50=1 μg/mL) than Escin or SIGMA saponin standard (IC50=5 μg/mL). See Figure 6 A.

Experiment 2: Determination the hemolytic and MTT activities of Compound Y after removal of the angeloly group or the sugar moiety by alkaline or acid hydrolysis, respectively Methods:

(A) Alkaline Hydrolysis of Xanifolia-Y: 20 mg of Xanifolia-Y was dissolved in 0.5 ml_ of 1 M NaOH. The solution was incubated in an 80 0 C water bath for 4 hours. It was cooled to room temperature before being neutralized with 0.5 ml_ 1 N HCI (adjusted 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 was further purified with HPLC in a C-18 column eluted with 25% acetonitrile.

(B) Acid Hydrolysis of Xanifolia-Y: 15 mg Xanifolia-Y was dissolved in 1 mL of Methanol. 1 mL of 2N HCI was then added. The mixture was refluxed in an 80 0 C water bath for 5 hours.

The solution was then neutralized by adding 2 mL of 1 N NaOH (to a final pH 3-4). The aglycone was then extracted with ethylacetate 3 mL x 3. The extracts were collected and

pooled. Further isolation of aglycone (sugar-removed Xanifolia-Y) was achieved by HPLC with isocratic elution of 80% acetonitrile. Results:

The angeloly groups or the sugar moiety of the compound Y were removed by alkaline or acid hydrolysis respectively. The hemolytic activities of the hydrolysed products were then analyzed. Results of these studies indicate that removing sugars from the compound Y reduced hemolytic activity, but removing the angeloyl groups from the compound Y destroyed the hemolytic activity. It also suggested that sugars are helpful but not essential for hemolytic activity. See Figure 4D. The experiment results show that compound-Y lost MTT activities if the angeloyl groups were removed. However, the MTT activities became very weak when the sugar moiety of the compound was removed. See Figure 5 C, 5 D. Results of comparison of hemoyltic activities between Compound Y, Escin from SIGMA are shown in Figure 7. Results of the comparison of hemolytic activities between compound Y, compound Y without sugar moiety or angeloly groups are shown in Figure 6 A, 6B. Chemical structures of compound Y without sugar moiety (ACH-Y) or angeloly groups (AKOH-Y) are shown in Figure 7 respectively.

Experiment 3: Effects of Xanifolia-Y on on reduction of venous insufficiency, particularly hemorrhoids (See PCT/US 2006/016158, filed April 27, 2006)

Experiment 4: Effects of Xanifolia Y on reduction of swelling of rats' feet in the Carrageenin -induced swollen feet model in rats.

(See PCT/US 2006/016158, filed April 27, 2006)

Experiment 5: Purification of the Inhibition Components in Extract.

(A) Fractionation of plant extracts with FPLC

(See PCT/US05/31900, filed Spetember 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. 11/131551 , filed May 17, 2005, the contents of which are incorporated herein by reference).

(B) Isolation of Component Ys with Preparative HPLC Methods

Column: A preparative HPLC column (Waters Delta Pak C18-300A); Elution conditions: 45% acetonitrile isocratic elution with flow rate of 1 ml/min. Fractions are monitored at 207nm and were collected and lyophilized.

Results

Final separation of Y fractions was achieved by HPLC with a preparative column. These fractions, which include compound YO, Y1 , Y2, Y (Y3) and Y4, were collected. Re- chromatography of compound Y showed a single peak in HPLC with a C18 reverse phase column. Re-chromatography of the compound Y7, Y8, Y9 and Y10 showed a single peak in HPLC with a C18 reverse phase column. (C) Appearance and solubility

The pure compound Ys is an amorphous white powder, soluble in aqueous alcohol, i.e., methanol or ethanol, 50% acetonitrile and 100% pyridine. (D) Inhibition analysis of Compound Ys with MTT assay

Inhibition analysis of compound Y was determined with MTT assay. Figure 5A shows the inhibition activities of compound Y, Y8, Y9 and Y10 on the growth of ovarian cancer cells (OCAR-3). Figure 27 shows the inhibition activities of compound Y's on the growth of ovarian cancer cells (OCAR-3)

Experiment 6: Determination of the Chemical Structure

Methods

NMR analysis. The pure compound Y of Xanthoceras sorbifolia was dissolved in pyridine-

D5 with 0.05% v/v TMS. All NMR spectra were acquired using a Bruker Avance 600 MHz NMR spectrometer with a QXI probe (1 H/13C/15N/31 P) at 298 K. The numbers of scans for 1 D 1 H spectra were 16 to 128, depending on the sample concentration. 2D HMQC spectra were recorded with spectral widths of 6000 x 24,000 Hz and data points of 2024 x 256 for t2 and t1 dimensions, respectively. The number of scans was 4 to 128. 2D HMBC were acquired with spectral widths of 6000 x 30,000 Hz and data points of 2024 x 512 for t2 and t1 dimensions, respectively. The number of scans was 64. The 2D data were zero-filled in t1 dimension to double the data points, multiplied by cosine-square-bell window functions in both t1 and t2 dimensions, and Fourier-transformed using software XWIN-NMR. The final real matrix sizes of these 2D spectra are 2048x256 and 2048x512 data points (F2χF1 ) for HMQC and HMBC, respectively. Mass spectral analysis. The mass of samples was analyzed by (A) MALDI-TOF Mass Spectrometry and by (B) ESI-MS Mass spectrometry. (A) Samples for MALDI-TOF were first dissolved in acetonitrile, and then mixed with the matrix CHCA, i.e., Alpha-cyano-4- hydroxycinnamic acid, 10mg CHCA/mL in 50:50 water/acetonitrile and 0.1 % TFA in final concentration. The molecular weight was determined by the high resolution mass spectroscope analysis with standards. (B) For ESI, the sample was analyzed with LCQ DECA XP Plus machine made by Thermo Finnigan. It is ionized with ESI source and the solvent for the compound is acetonitrile.

Results

The profile of the proton NMR is presented in Figure 8A, 8B, 9A, 9B, 10A, 10B and. The NMR profiles of 1 H, 13C, TOCSY, HMQC, HMBC and NOESY are shown respectively. Based on these data and analysis, the structure of compound YO is assigned as shown below.

Structure of Compound YO

This invention provides a bioactive compound YO and the chemical name is: 3-O-[β-D-galactopyranosyl(1→2)]-α-L-arabinofuranosyl(1↠’3)-β-D-glucuronopyranosyl-21- O-angeloyl, 22-O-(2-methylpropanoyl)-3β, 15α, 16α, 21 β, 22α, 28-hexahydroxyolean-12- ene,

Experiment 7: Animal Study • Methods

• Athymic Nu/Nu mice are divided into three groups (A, B and C) with four animals in each group.

• On day 0, mice of group A and B were transplanted intra-peritoneally with ES2 (human ovarian cancer) cells. • On day 1 , mice from B and C groups received drug (Xanifolia-Y, by i.p. route at dose of 5 mg/kg)

• On days 2 to 4, and 7 to 11 , B and C groups animals received daily drug administration of Xanifolia-Y, by i.p. route at dose of 2.5 mg/kg.

• Group A mice have no drug-treatment. Results:

Group A Mice - Died on day 19-22; Group B Mice - Survived over 50 days; Group C Mice - Survived over 50 days; Also See Figure 21 Experiment 8: Animal Study

• Methods • Athymic Nu/Nu mice were divided into three groups (A, D and E) with four animals in each group.

• On day 0, all mice were transplanted intra-peritoneally with ES2 (human ovarian cancer) cells.

• Group A mice received no drug-treatment.

• Group D: From day 4, mice received a daily drug administration of Xanifolia-Y, via i.p. route for 9 days at dose of 2.5 mg/kg.

• Group E: From day 10, mice received daily drug administration of Xanifolia-Y, via i.p. route for 10 days at dose of 2.5 mg/kg.

Result:

Group A, Mice implanted with tumor and no drug. All died within 24 days; Group D, Mice implanted with tumor and were given drug 9 times from 4th day. All survived; Group E Mice implanted with tumor and were given drug 10 times from 10th day. Half the number of mice survived. Also See Figure 22

Experiment 9: Animal Study • Athymic Nu/Nu mice (2-3 months old) were transplanted sc with ES2 (human ovarian cancer) cells.

• Five days after the transplant (day one), mice were divided into two groups (H and J) with two animals in each group.

• Group H: On days 1-5, and 8-10 mice received daily drug administration of Xanifolia- Y, by i.p. route at dose of 2.5 mg/kg.

• Group J mice received no drug-treatment. Result:

Group H: Mice received drug-treatment, tumor size is 10 mm in 10 days Group J: Mice received no drug-treatment, tumor size is 18 mm in 10 days The tumor size is 45% smaller in mice with drug than the mice with no drug in 10 days period. See Figure 23

Experiment 10: Aminal Study • Methods

• Athymic Nu/Nu mice (5-6 weeks old) are divided into three groups (O, P and Q) with 5-6 animals in each group.

• On day 0, all mice were transplanted intra-peritoneally with ES2 (human ovarian cancer) cells. • Group O: animals received no drug-treatment.

• Group P: On days 4-8, 11-15, 18-22, 25-29, 32-36,39-43, animals received daily drug administration of Xanifolia-Y, by i.p. route at dosage of 2.5 mg/kg

• Group Q: On days 10-15, 18-22, 25-29, 32-36, 39-43, animals received daily drug administration of Xanifolia-Y, by i.p. route at dosage of 2.5 mg/kg. Result:

The median survival time of tumor bearing mice without drug-treatment is 24 days. The median survival time of tumor bearing mice with drug-treatment starting on day 4 after tumor inoculation is 58 days (extension of life span of 141 %); and The median survival time of tumor bearing mice with drug-treatment started on day 10 after tumor inoculation is 31 days (extension of life span of 29%). See Figure 28

Experiment 11 : Studies of effect on human ovarian cancer cell lines.

Since we found that ovarian carcinoma cell lines are among the sensitive cells studied, we further investigated if other ovarian carcinoma cell lines are also susceptible to Xanifolia-Y. Majority of ovarian cancers arise from the surface epithelium of ovary, most of them belong to the histological subtypes of clear cell and serous carcinoma. We obtained 10 more human ovarian carcinoma cell-lines of these histological subtypes for these studies. The inhibition activity exerted by Xanifolia-Y was determined with MTT assay. The following table shows the IC50 values of Xanifolia-Y on these cell lines. Results: Table 2. IC50 values of human ovarian carcinoma determined by MTT assay.

The IC50 values of Xanifolia-Y in these cell-lines are ranging from 2 to 10 uM. These studies show that the effective concentration of Xanifolia-Y is in the micro-molar range which is comparable to those of other anti-cancer drugs.

Experiment 12: Study apoptosis induced by Xanifolia-Y.

Experiment of apoptosis of OVCAR3 cells after treatment with Xanifolia-Y was assessed with flow cytometry of GFP-Annexin-V and propidium iodide.

Results were shown in Fig. 25 which indicates that induction of the early apoptosis (the lower right quadrant) and late a poptotic/necrosis (the upper right quadrant) were found in cells 24 h after exposure to Xanifolia-Y. By comparing the distribution of apoptotic/necrotic cells after the drug-treatment, a higher number of early apoptotic cells were observed as compare to those of the late apoptotic/necrosis cells. These results indicate that apoptosis is a major form of cell death induced by Xanifolia-Y. See Figure 24.

Experiment 13: Effect of Xanifolia-Y on membrane structure (EM studies). Xanifolia-Y has a potent hemolytic activity in red blood cells (Fig. 26). To study the effect of

Xanifolia-Y on membrane structure, the morphology of cell membrane treated with Xanifolia-Y was examined with EM. In this experiment, K562 cells were treated with 5 uM of Xanifolia-Y for 60 min. Solvent DMSO and AKOH-Y (a derivative of Xanifolia-Y without the angeloyl group and it has no activity) served as controls. Cells were negative stained with 1 % UAc and subsequently examined with EM.

Figure 25 show that patches of pits were found in the membrane of Xanifolia-Y treated cells (Fig. 25B) but not in cells treated with the DMSO (Fig. 25A) or AKOH-Y (Fig. 25C) controls. These pits have the size from 8OA to 500A (in diameter). The pits represent holes formed in the membrane. The pits are arranged in a characteristic pattern with smaller pits (8OA in diameter) located in the periphery and the bigger ones (500A in diameter) in the center. The bigger holes are resulted from fusing of the smaller holes (Fig. 25D).

Membrane image of cells treated with A: DMSO solvent control, 60 min (magnification: X60,000); B: Xanifolia-Y 5 uM, 60 min. (X60000); C: AKOH-Y, 20 uM, 60 min. (X60000); D: Xanifolia-Y 5 uM, 60 min. (X20000)

This experiments results show that the Xanifolia-Y alters the membrane of cell.

Experiment 14: Inhibition of cell adhesion by Xanifolia-Y.

Methods and Results. ES2 or HeyδA cells were plated in T25 flasks with medium containing 5 ug/ml of Xanifolia-Y. Cultures were incubated for 5 hours. Attached cells were removed from flasks by trypsinization and the amounts were counted. Compare to no drug controls, 86 ± 4 % of ES2 cells and 67 ± 8 % of HeyδA cells were found attached to flasks under this condition. At 5 ug/ml Xanifolia-Y, over 90% of unattached cells are alive as determined by the trypan Blue exclusion assay and by their ability to re-attach to flasks when plating in medium without Xanifolia-Y. However, with 10 ug/ml Xanifolia-Y, less than 40% of cells attached to flasks and many of them are dead cells. This experiment shows that Xanifolia-Y inhibits cells adhesion process.

Experiment 15: Combined inhibition effect of Xanifolia-Y and Paclitaxel.

Methods: ES2 cells were exposed to (i) Xaniffolia-Y with concentrations of 40, 20, 10, 5, 2.5, and 1.25 ug/ml; or (ii) Paclitaxel with concentrations of 10, 5, 2.5, 1.25, 0.62 and 0.031 ng/ml; or (iii) Combined Xanifolia-Y and Paclitaxel with concentrations of each drug in same order (for example, 40 ug/ml Y plus 10 ng/ml T; 20 ug/ml Y plus 5 ng/ml T, etc.). Cells growth under these conditions was determined by the MTT assay.

Results: The IC50 for Xanifolia-Y and Paclitaxel, is 5ug/ml and 1.25 ng/ml, respectively. Additive effect was observed when both drugs were used, because in this case, the IC50 value for Paclitaxel (0.625 ng/ml) and for Xanifolia-Y (2.5 ug/ml) is less than those when they are used singly. See Figure 26.

Experiment 18: Determination of Aquaporin in HeLa and OVCAR3 cells. Methods: 1. HeIa or OVCAR3 cells were cultured in RPMI 1640 medium at 37C in an incubator with 5% CO2.

2. Cells were harvested, washed with PBS.

3. Cellular protein was dissolved in SDS sample buffer with protease inhibitors (PMSF and Leupeptin) and was incubated at 7OC for 20 min before use. 4. Equal amounts of protein from HeIa or OVCAR3 cells were separated with 12% SDS gel and subsequently blotted on nitrocellulose paper.

5. Western blot was performed with anti-AQ1 antibody (Chemicon/SIGMA) and second antibody which was conjugated with Alkaline phosphatase.

Results: The following figure shows the results of Western blot.

Aquaporin-1 (indicated with an arrow) was observed in OVCAR3 cells but was minimally detected in HeLa cells.

Based on same amounts of protein loading into gel, it was found that OVCAR3 cells have higher concentration of Aquaporin-1 than in HeIa cells. Since OVCAR3 cells are more sensitive to Xanifolia-Y and it has a higher concentration of

Aquaporin-1 , these results show Xanifolia-Y is potent to inhibit the cancer cell growth wherein the Aquaporin is overexpressed.

See Figure 31

References

1. Voutquenne, L., Cecile Kokougan. Catherine Lavaud, lsabelle Pouny, Marc Litaudon. Triterpenoid saponins and Acylated prosapogenins from Harpullia austro-caledonica." Phytochemistry 59 (2002) 825-832. 2. Zhong Jaing, Jean-francois Gallard, Marie-Therese Adeline, Vincent Dumontet, Mai Van Tri, Thierry Sevenet, and Mary Pais "Six Triterpennoid Saponins from Maesa laxiflora." J. Nat. Prod. (1999), 62, 873-876.

3. Young Seo, John M. Berger, Jennine Hoch, Kim M Neddermann, lsia Bursuker, Steven W. Mamber and David G. Kingston. "A new Triterpene Saponin from Pittosporum viήdiflorum from the Madagascar Rainforest ". J. Nat. Prod. 2002, 65, 65-68.

4. Xiu-Wei Yang, Jing Zhao, Xue-Hui Lui, Chao-Mei Ma, Masao Hattori, and Li He Zhang "Anti-HIV-1 Protease Triterpenoid Saponins from the Seeds of Aesculus chinensis." J. Nat. Prod. (1999), 62, 1510-1513.

5. Yi Lu, Tatsuya Umeda, Akihito Yagi, Kanzo Sakata, Tirthankar Chaudhuri, D. K. Ganguly, Secion Sarma. "Triterpenoid Saponins from the roots of the tea plant (Camellia sinensis var. Assamica)." Phytochchemistry 53 (2000) 941-946.

6. Sandra Apers, Tess E. De Bruyne, Magda Claeys, Arnold J. Viletinck, Luc A.C. Pieters. "New acylated triterpenoid saponins from Maesa laceceolata." Phytochemistry 52 (1999) 1121-1131. 7. Maria D'Acquarica, Maria Cristina, Di Giovanni, Francesco Gasparrini, Domenico Misiti, Claudio D' Arrigo, Nicolina Fagnano, Decimo Guarnieri, Giovanni lacono, Giuseppe Bifulco and Raffaele Riccio. "Isolation and structure elucidation of four new triterpenoid estersaponins from fruits of the Pittosporumtobira AIT." Tetrahedron 58 (2002) 10127- 10136. 8. Method for prevention and treatment of chronic venous insufficiency, Jia et al., U.S.

Patent No.6210680

9. Novel analgestic compounds, extracts containing same and methods of preparation. Quinn Ronald, WO 2005/051969