US20060110469A1 - Angelicae sinensis extracts useful for treatment of cancers - Google Patents

Angelicae sinensis extracts useful for treatment of cancers Download PDF

Info

Publication number
US20060110469A1
US20060110469A1 US11/246,009 US24600905A US2006110469A1 US 20060110469 A1 US20060110469 A1 US 20060110469A1 US 24600905 A US24600905 A US 24600905A US 2006110469 A1 US2006110469 A1 US 2006110469A1
Authority
US
United States
Prior art keywords
extract
angelicae
sinenses
subject
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/246,009
Inventor
Jiann-Kuan Luo
Horng-Jyh Harn
Wen-Liang Chang
Shinn-Zong Lin
Yeung-Leung Cheng
Nu-Man Tsai
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Buddhist Tzu Chi General Hospital
Original Assignee
Buddhist Tzu Chi General Hospital
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Buddhist Tzu Chi General Hospital filed Critical Buddhist Tzu Chi General Hospital
Priority to US11/246,009 priority Critical patent/US20060110469A1/en
Assigned to BUDDHIST TZU CHI GENERAL HOSPITAL reassignment BUDDHIST TZU CHI GENERAL HOSPITAL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIN, SHINN-ZONG, LUO, JIANN-KUAN, TSAI, NU-MAN, HARN, HORNG-JHY, CHENG, YEUNG-LEUNG, CHANG, WEN-LIANG
Assigned to BUDDHIST TZU CHI GENERAL HOSPITAL reassignment BUDDHIST TZU CHI GENERAL HOSPITAL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NULIV SCIENCE USA, INC.
Publication of US20060110469A1 publication Critical patent/US20060110469A1/en
Priority to US11/561,713 priority patent/US7455861B2/en
Priority to US12/260,723 priority patent/US20090111870A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/23Apiaceae or Umbelliferae (Carrot family), e.g. dill, chervil, coriander or cumin
    • A61K36/232Angelica
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention mainly relates to a new use of an acetone extract, chloroform extract or hexane extract of Angelicae sinesis and the active components purified therefrom in the treatment of cancers.
  • Cancers are abnormal cell proliferations that result from the accumulation of genetic changes in cells endowed with proliferative potential. Treatment of cancers has relied mainly on surgery, chemotherapy, radiotherapy and more recently immunotherapy. However, new approaches for treating and preventing cancers are still desired.
  • Angelicae sinensis (Dangqui) is one of the most frequently occurring drugs in the prescriptions of traditional Chinese medicines.
  • the traditional uses of Angelicae sinensis include those to promote blood production, protect liver, lower blood pressure, kill bacteria, ease pain mostly for menstrual disorder in women, and lower cholesterol (Chinese Herbs, Shanghai Science and Technology Publication, Inc., Shanghai, China, Vol. 5, p. 893, 1999).
  • CN1053747 disclosed that Angelicae sinensis (Oliv) Diels, ASD, and the ASDP and ASDE as effective components of an adjuvant was prepared, and could be used as an immunological adjuvant to genetically-engineered hepatitis B vaccines. It was reported in CN1109356 that the effective component, lactones (ASDE), extracted from Angelicae sinensis (oliv) diels, ASD, could be used as an immunological adjuvant, which can enhance the immunogenicity and help lower toxicity. Kumazawa et al.
  • This invention provides that an acetone extract, chloroform extract or hexane extract of Angelicae sinensis, or at least one component purified therefrom, such as n-butylidenephthalide (BP), can also inhibit telomerase activity of cancer cells and further induce their apoptosis so that they can be used to treat malignant neoplasms. Therefore, an acetone extract, chloroform extract or hexane extract of Angelicae sinenses, and the components purified therefrom, such as n-butylidenephthalide, are potent for manufacturing of medicines for the treatment of cancers, and can be used in combination with chemotherapy drugs through their activities on cell cycle regulation, and telomerase inhibition.
  • BP n-butylidenephthalide
  • one object of the present invention is to provide a method for inhibiting cancer cell proliferation and migration in tumor tissues.
  • Another object of the present invention is to provide a method for inhibiting telomerase activity of cancer cells.
  • Yet another object of the present invention is to provide a method for inducing apoptosis of cancer cells.
  • Another object of the present invention is to provide the use of an acetone extract, chloroform extract or hexane extract of Angelicae sinenses, or at least one component purified therefrom, such as n-butylidenephthalide, for manufacturing medicine for the treatment of cancer, and as an adjuvant in combination with chemotherapy drugs through their activities on cell cycle regulation and/or telomerase inhibition.
  • FIG. 1 a provides the results of the cell cycle analysis, which demonstrates that treatment with 70 ⁇ g/ml AS-C (the chloroform extract of Angelicae sinensis ) enhanced cell cycle accumulation at G0/G1 phase (>90%) in GBM cells (DBTRG-05M) (*p ⁇ 0.05) with a concurrent decrease in S phase.
  • AS-C the chloroform extract of Angelicae sinensis
  • FIG. 1 b provides the results of the cell cycle analysis, which demonstrates that the treatment with 400 ⁇ M BP enhanced cell cycle accumulation at G0/G1 phase (>90%) in GBM cells (DBTRG-05MG) (*p ⁇ 0.05, **p ⁇ 0.005) with concurrent decrease in S phase.
  • FIG. 2 shows the effect of BP (n-butylidenephthalide) of 5 to 800 ⁇ M, in inducing GBM tumor cell (DBTRG-05MG) apoptosis, as assessed by TUNEL method, using propidium iodide as a counter staining (*p ⁇ 0.05, ** p ⁇ 0.005, *** p ⁇ 0.0005).
  • BP n-butylidenephthalide
  • FIGS. 3 a - 3 c provide the results of the analyses of apoptosis pathways induced by AS-C, 70 ⁇ g/ml (wherein the DBTRG-05MG cell line was used.)
  • FIGS. 3 d - 3 g provide the results of the analyses of apoptosis pathways induced by BP, 400 ⁇ M (wherein the DBTRG-05MG cell line was used.)
  • FIG. 4 shows the inhibitory effect of the AS-C treatment (500 mg/kg) on the tumor sizes in mice bearing subcutaneous GBM tumors (RG-2) (p ⁇ 0.05).
  • FIG. 5 shows that the survival rate of the AS-C treated mice (dose—500 mg/kg) was significantly prolonged as compared with the control group (p ⁇ 0.0001), wherein the DBTRG-05MG cell line was used.
  • FIG. 6 shows the inhibitory effect of AS-C treatment (500 mg/kg) on the growth of in situ GBM tumor (RG2) volume on rats (* p ⁇ 0.05, ** p ⁇ 0.001).
  • FIG. 7 shows the inhibitory effect of the AS-C treatment (500 mg/kg by intra-peritoneal or subcutaneous administration) on the xenograft tumor growth of mice (p ⁇ 0.005), wherein the DBTRG-05MG cell line was used.
  • FIG. 8 shows the inhibitory effect of the BP treatment (300 mg/kg) on tumor volume of GBM in situ (RG2) on rats, which was calculated with MRI imaging using echo-planar imaging capability (* p ⁇ 0.05, ** p ⁇ 0.001).
  • FIG. 9 shows the inhibitory effect of BP treatment, at different dosages (70 to 800 mg/kg), on xenograft tumor growth on mice (p ⁇ 0.005), wherein the DBTRG-05MG cell line was used.
  • FIG. 10 shows the effect of BP treatment (70 to 800 mg/kg) on the prolongation of survival period of nude mice with xenograft tumor (subcutaneous DBTRG-05MG) (p ⁇ 0.001).
  • This invention provides that the organic solvent extracts of Angelicae sinensis, or the components purified therefrom, such as n-butylidenephthalide (BP), can inhibit telomerase activity of cancer cells and further induce their apoptosis. Therefore, they can inhibit cancer cell proliferation and can be used to treat cancers.
  • BP n-butylidenephthalide
  • Angelicae sinensis has long been used in blood diseases and female diseases. Normally, the dried root of Angelica sinensis (Oliv.) Diels, belonging to the family of Umbelliferae, is used. Angelica sinensis (AS) is appreciated by those skilled in this art. A variety of techniques are well known in the art for extracting, separating, and/or purifying individual active components of Angelicae sinensis. The organic solvent extracts of Angelicae sinensis may be obtained by any standard procedures commonly used in the field. According to the invention, Angelicae sinensis is extracted with acetone, chloroform, or hexanes.
  • the dried and powdered rhizomes of Angelicae sinensis were extracted with acetone as a solvent to give an extract as AS-A. Furthermore, AS-A was further extracted with chloroform to give an extract as AS-C; and AS-A was further extracted with hexanes to give an extract as AS-H.
  • Active components of Angelicae sinensis may be isolated and/or purified from the organic solvent extracts of Angelicae sinensis by using any techniques known in the art.
  • the active components may be purified from Angelicae sinensis in any form, particularly the rhizomes.
  • Various techniques that may be employed in the further purification include filtration, selective precipitation, extraction with organic solvents, extraction with aqueous solvents, column chromatography, high performance liquid chromatography (HPLC), etc.
  • some active components were purified from the organic solvent extracts of Angelicae sinensis, such as ligustilide and n-butylidenephthalide, which can induce tumor cell apoptosis.
  • E- and Z-geometrical isomers of n-butylidenephthalide (BP) were separated with column chromatography and characterized with HPLC and NMR.
  • Telomeres the extremities of eukaryotic chromosomes, are essential for maintaining the integrity of the genome and are a key determinant of cellular aging and immortality (N. W. Kim, M. A. Piatyszek, K. R. Prowse, C. B. Harley, M. D. West, P. L. C. Ho, G. M. Coviello, W. E. Wright, S. L. Weinrich, J. W. Shay, Science, 266, 2011-2015 (1994)). Telomere length and the rate of its reduction vary among organs and individuals.
  • telomere length regulators G. B. Morin, Cell, 59, 521-529 (1989)
  • Apoptosis is another mechanism of cancer therapy, which has become one of the newest areas of cell biology research.
  • apoptosis program The activation of the apoptosis program is regulated by various signals from both intracellular and extracellular stimuli. Indeed, in recent years evidence is beginning to accumulate that many (and perhaps all) agents of cancer chemotherapy kill tumour cells by launching the mechanisms of apoptosis. New drugs associated with apoptosis are expected to be most effective against tumours with high proliferation rates. Many such candidates are being screened for use in the treatment of cancer (Ricardo Pérez-Tovon, Beatriz Montaner, Esther Llagostera, kann Soto-Cerrato, Biochemical Pharmacology, 66, 1447-1452 (2003)).
  • Apoptosis is monitored by the analysis of two commonly used endpoints—the morphological changes of cells (condensation of nuclear chromatin, formation of apoptotic bodies) and DNA fragmentation into large fragments (300 and 50 kbp) and then to oligonucleosomesized fragments (multiples of 200 bp), which appear as a “ladder” of DNA bands upon agarose gel electrophoresis.
  • endpoints is an indicator of apoptosis, quantification of the percentage of apoptotic cells in a population by such an assay is impossible.
  • the relative contribution of the receptor and the mitochondrial pathways to drug-induced apoptosis has been a subject of controversy. It depends on the type of the cytotoxic drug itself, the dose and kinetcs or on differences between certain cell types, which affects the cell type dependent signaling in the Fas/FasL pathway.
  • Apoptosis pathways can be initiated through different sites, such as the plasma membrane, by way of death receptor mediated signaling (receptor pathway; Fas/FasL/caspase-8/caspase-3 pathway), the mitochondria (mitochondrial pathway; Bax/AIF/caspase-9/caspase-3 pathway), and cell cycle regulation (including p53, Rb tumor suppressors, p16 and p21 cyclin kinase inhibitors and cyclin/cdk cell cycle check points) (Simone Fulda, Matroulea, Klaus-Micheal Debatin, Cancer Letter, 197, 131-135 (2003)).
  • death receptor mediated signaling receptor pathway; Fas/FasL/caspase-8/caspase-3 pathway
  • mitochondria mitochondria
  • Bax/AIF/caspase-9/caspase-3 pathway mitochondria
  • cell cycle regulation including p53, Rb tumor suppressors, p16 and p21 cyclin kinase inhibitors and
  • the growth of several cancer cell lines were tested against the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, the active components purified therefrom, such as n-butylidenephthalide. It was found that they were cytotoxic to cancer cells; they inhibited telomerase activity of cancer cells (as shown in Example 7); they suppressed cancer cell proliferation (as shown in Example 2) and they also induced cancer cell apoptosis (as shown in Examples 3 and 4). Furthermore, animal studies also showed they were effective in suppressing cancer growth (as shown in Examples 5 and 6). They are therefore potent for treating cancers, particularly human malignant glioblastoma, colorectal cancer, leukemia, neuroblastoma, hepatoma, breast, ovarian and lung cancers.
  • the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives according to the present invention may be administered by any conventional route of administration including, but not limited to, oral, parenteral, intraperitoneal (ip), intravenous (iv), intramuscular (im), subcutaneous (sc), pulmonary, transdermal, buccal, nasal, sublingual, ocular, rectal, vaginal or other routes. It will be readily apparent to those skilled in the art that any dosage or frequency of administration that provides the desired therapeutic effect is suitable for use in the present invention. In a preferred embodiment of the invention, they are administered by oral delivery, using methods known to those skilled in the art of drug or food delivery.
  • the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives may be in the form of a tablet, pill, capsule, granule, gel, powder, sterile parenteral solution or suspension, metered aerosol or liquid spray, or suppository, depending on the administration route.
  • the organic solvent extracts of Angelicae sinensis, active components purified therefrom, or the derivatives are admixed with a pharmaceutically acceptable carrier according to conventional pharmaceutical compounding techniques, wherein the carrier may take a wide variety of forms depending on the form of preparation desired for administration. Suitable pharmaceutically acceptable carriers are well known in the art.
  • the tablets, capsules, gels, solutions or suspensions may also include the following components: a pharmaceutically acceptable excipient or carrier, which is a non-toxic, inert solid or semi-solid, diluent, encapsulating material, a gel base or formulation auxiliary of any type.
  • the solutions and suspensions may contain auxiliaries, such as water for injection, saline solution, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; proteins such as serum albumin to enhance solubility; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • auxiliaries such as water for injection, saline solution, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; proteins such as serum albumin to enhance solubility; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives according to the present invention may also be administered as an adjuvant in combination with chemotherapy drugs, such as actinomycin, adriamycin, Ara-C, bleomycin, carmustin, cisplatin, cyclophosphamide, daunomycin, mitomycin, taxol, vinblastine, etc.
  • chemotherapy drugs such as actinomycin, adriamycin, Ara-C, bleomycin, carmustin, cisplatin, cyclophosphamide, daunomycin, mitomycin, taxol, vinblastine, etc.
  • acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives according to the present invention may also be formulated in any dietary compositions by using any techniques known to those skilled in the art.
  • the roots of Angelicae sinensis were supplied from Chung-Yuan Co., Taipei, Taiwan and were identified by Professor Han-Ching Lin. A voucher of herbarium specimen was deposited at the School of Pharmacy, National Defense Medical Center.
  • the dried and powdered rhizomes of Angelicae sinensis (12 kg) were extracted 3 times with acetone (24 L/time) to give an acetone extract called AS-A.
  • AS-A was then subjected to chloroform extraction 3 times (24 L/times). The latter extracts were concentrated under reduced pressure to yield 31.67 g of chloroform extract, called AS-C (from 100 g of acetone extract).
  • hexane extract was obtained by extracting AS-A with hexanes.
  • n-Butylidenephthalide (BP) was purchased from Lancaster Synthesis Ltd. (Newgate, Morecambe, UK) and used without further purification. E- and Z-forms of BP were separated with column chromatography and characterized with HPLC and NMR. They were dissolved in DMSO, incubated with shaking at 25° C. for 1 hour and stored at 4° C. before each in vitro experiment.
  • Human umbilical vascular endothelial cells were purchased from Cascade Biologics, Inc. (USA). HUVECs were maintained in Medium 200 (Cascade Biologics, Inc. USA) supplemented with 10% fetal bovine serum (FBS; Gibco BRL) and low serum growth supplement (LSGS; Cascade Biologics, Inc. USA). Human dermal fibroblasts (HDFs) were purchased from Cascade Biologics, Inc. (USA). HDFs were maintained in Medium 106 (Cascade Biologics, Inc. USA) supplemented with 10% FBS and low serum growth supplement (LSGS; Cascade Biologics, Inc. USA).
  • FBS fetal bovine serum
  • LSGS low serum growth supplement
  • HDFs Human dermal fibroblasts
  • HDFs were purchased from Cascade Biologics, Inc. (USA). HDFs were maintained in Medium 106 (Cascade Biologics, Inc. USA) supplemented with 10% FBS and low serum growth supplement (LSGS;
  • the human colon adenocarcinoma cell lines HT-29 were purchased from the ATCC (Manassas, Va. USA). HT-29 were maintained in Dubecco's Modified Eagle Medium (DMEM; Gibco) supplemented with 10% FBS and 100 ng/ml penicillin and 100 ng/ml streptomycin (Life Technologies, Inc., Grand Island, N.Y., USA). For the proliferation assay of cell in log growth (G1), the cells were harvested at 60-80% of confluence.
  • DMEM Dubecco's Modified Eagle Medium
  • the cells were lysed directly in a 6 well culture plate by adding 1 ml of trizol reagent to a 3.5 cm diameter well, and passing the cell lysate several times through a pipette.
  • Phase separation The homogenized samples were incubated for 5 minutes at 15 to 30° C. to permit the complete dissociation of nucleoprotein complexes. Then, 0.2 ml of chloroform (Riedel-de-ha ⁇ n) per 1 ml of trizol reagent was added. Sample tubes were capped securely. The tubes were vigorously shaken by hand for 15 seconds and incubated at 15 to 30° C. for 2 to 3 minutes. The samples were centrifuged at no more than 12,000 ⁇ g for 15 minutes at 2 to 8° C. Following centrifugation, the mixture separated into a lower red, phenol-chloroform phase, an interphase, and a colorless upper aqueous phase. RNA remained exclusively in the aqueous phase.
  • RNA precipitation The aqueous phase was transferred to a fresh tube. The RNA was precipitated from the aqueous phase by mixing with isopropyl alcohol (Fluka). 0.5 ml of isopropyl alcohol was added for every 1 ml of trizol reagent which was added for homogenization initially. The samples were incubated at 15 to 30° C. for 10 minutes and centrifuged at no more than 12,000 ⁇ g for 10 minutes at 2 to 8° C.
  • isopropyl alcohol Fluka
  • 0.5 ml of isopropyl alcohol was added for every 1 ml of trizol reagent which was added for homogenization initially. The samples were incubated at 15 to 30° C. for 10 minutes and centrifuged at no more than 12,000 ⁇ g for 10 minutes at 2 to 8° C.
  • RNA wash The supernatant was removed. The RNA pellet was washed once with 75% ethanol, using at least 1 ml of 75% ethanol for every 1 ml of trizol reagent, which was added for the homogenization initially. The sample was mixed by vortexing and centrifuged at no more than 7,500 ⁇ g for 5 minutes at 2 to 8° C.
  • RNA Re-dissolving the RNA: The supernatant was removed, and then the RNA pellet was dried. The RNA was dissolved in RNase-free water, and incubated for 10 minutes at 55 to 60° C. The RNA can be stored at ⁇ 70° C.
  • Human tumor cell lines (MCF-7, CL1-5, HT-29, Caco-2), human umbilical vascular endothelial cells (HUVEC), and human dermal fibroblasts (HDF) were tested for sensitivity on Angelicae sinensis extracts, ligustilide, n-butylidenephthalide and its derivatives in vitro.
  • the DBTRG-05MG, BCM, HL-60 and J5 cells were grown in RPMI-1640 medium containing 10% fetal calf serum and 100 ng/ml penicillin and 100 ng/ml streptomycin at 37° C. in a humidified atmosphere with 5% CO 2 .
  • the G5T/VGH, RG2, N18, SVEC and Balb/3T3 cells were cultured in DMEM with 10% fetal calf serum and 100 ng/ml penicillin and 100 ng/ml streptomycin at 37° C. in 5% CO 2 .
  • Mycoplasma infection of the culture cells was excluded by PCR screening methods before each experiment.
  • the effects on cell viability after the treatments with different concentrations of the Angelicae sinensis extracts or the active components purified therefrom were evaluated by modified MTT assay in triplicate. Briefly, the cells (5 ⁇ 10 3 ) were incubated into 96-well plates containing 100 ⁇ l of a growth medium. The cells were permitted to adhere for 24 hours, then treated with 100 ⁇ l of the herbal extracts or the active components dissolved in the medium. The control contained DMSO of ⁇ 0.02% (v/v). After 24, 48 and 72 h incubation, the drug-containing medium was replaced by 50 ⁇ l of fresh medium, and cells in each well were incubated in 50 ⁇ l of 400 ⁇ g/ml MTT for 6-8 h.
  • BP n-butylidenephthalide
  • AS-A Angelicae sinensis extracts
  • AS-C Angelicae sinensis extracts
  • the IC 50 of AS-C and BP to brain tumor cell lines were 35 ⁇ 60 ⁇ g/ml and 1.4 ⁇ 10 ⁇ g/ml, respectively, while those to the normal cell line (HDF) were 85 ⁇ 300 ⁇ g/ml, (p ⁇ 0.0001).
  • vascular endothelial cells 44.2 ⁇ 0.1 ⁇ g/ml
  • the GBM cells After treatment with AS-C or BP, the GBM cells (DBTRG-05MG) were seen to be detached and floating in the media at different points of time within a 72-hour period of observation. The extent of GBM cell detachment and flotation was found to increase with time, and with the increase in dosage (in the case of BP when an observation was made at 3 hours).
  • the GBM cell detachment and flotation after treatment with AS-C or BP can be attributed to morphology change in the tumor cells.
  • BCNU carmustin
  • Brain tumor cell lines DBTRG-05MG and G5T/VGH were cultured in the growth medium with a diluent.
  • DMSO was added, and the content is less than 0.02% (v/v).
  • AS-C and BP treatment 70 ⁇ g/ml of AS-C and 400 ⁇ M of BP were added, respectively. All were cultured for 48 hours.
  • the analysis of cell cycle distribution was performed by DNA staining with propidium iodide (PI). Briefly, 2 ⁇ 10 6 adherent cells were detached by trypsinization. The detached cells and the floating dead cells were centrifuged and washed twice with 10 ml of cold 1 ⁇ PBS (Life Technologies, Inc.).
  • the G 0 /G 1 phases were gated in M1 ( ⁇ 2); G2/M phases were gated in M2 ( ⁇ 2); the total cells were gated in M3; S phase was M3 ⁇ (M1( ⁇ 2)+M2( ⁇ 2)); Sub G1 phase (apoptosis cells) was gated in M4.
  • FIGS. 1 a and 1 b showed that both AS-C and BP enhanced a significant G0/G1 phase arrest with a concurrent decrease of S phase after treatment for 12 hours to 48 hours (p ⁇ 0.05,p ⁇ 0.005).
  • Apoptotic cell death was analyzed using In Situ Cell Death Detection Kit, POD (Roche, Germany). Changes in DNA chromatin morphologic features were used for quantification. The procedures were performed in accordance with the manufacturer's instructions. Briefly, cells were cultured on culture dish and analyzed 72 hours after treatment with AS-C (70 ⁇ g/ml) and BP (5 ⁇ 800 ⁇ g/ml), respectively. In AS-C and BP-treated groups, the suspended cells were collected. In the control group, adherent and floating cells were collected. Then, the cells were fixed with 3.7% formaldehyde at room temperature for 15 min.
  • DBTRG-05MG cells human GBM cells
  • AS-C 70 ⁇ g/ml
  • DBTRG-05MG cells were treated with BP (400 ⁇ M) for 0, 1.5, 3, 6, 12, 24 and 48 hours.
  • the cell pellets were re-suspended in lysis buffer (10 nM Tris-HCl, pH 7.5, 1 mM EGTA, 0.5% CHAPS, 10% (v/v) glycerol, 5 mM , ⁇ -2-mercaptoethanol and 0.1 mM phenylmethylsulfonyl fluoride) and incubated on ice for 30 min, and then centrifuged at 13000 ⁇ rpm at 4° C.
  • lysis buffer 10 nM Tris-HCl, pH 7.5, 1 mM EGTA, 0.5% CHAPS, 10% (v/v) glycerol, 5 mM , ⁇ -2-mercaptoethanol and 0.1 mM phenylmethylsulfonyl fluoride
  • the protein concentration of whole cell lysates was measured with BCA protein assay kit (Pierce, Rockford, Ill.) following the manufacturer's instructions.
  • the cell lysates (20 ⁇ g/lane) were electrophoresed on 10-12% SDS-PAGE (Bio-Rad, Hercules, Calif.). Proteins were transferred to polyvinyldenefluoride (PVDF) membranes (Amersham Lifesciences, Piscataway, N.J.).
  • PVDF polyvinyldenefluoride
  • the membranes were masked for 1 hour at room temperature with 5% skim milk as the blocking agent, and incubated with the respective antibodies of Fas (FL-335), Fas-L (C-178), caspase 3 (H-277), caspase 8 (H-134), caspase 9 (H-170), Bax (B-9), p16 (F-12), p21 (F-5), p53 (DO-1; 1/100 dilution) (Santa Cruz Biotechnology Inc., Calif., U.S.A.), phospho-p53 (Ser15; 1/2000 dilution) and phospho-Rb (Ser795; 1/2000 dilution) (Cell Signaling Technology, Mass., USA) for 2 hours at room temperature.
  • Fas FL-335
  • Fas-L C-178
  • caspase 3 H-277
  • caspase 8 H-134
  • caspase 9 H-170
  • Bax B-9
  • p16 F-12
  • p21 F-5
  • p53
  • Antibody recognition was detected, by incubating the membranes with the respective anti-mouse, anti-rabbit, anti-goat IgG secondary antibodies ( 1/1000 dilution; Santa Cruz Biotechnology Inc., Calif., U.S.A.) conjugated to horseradish peroxidase, for 1 hour at room temperature, and visualized with the ECL Plus chemiluminescence system (Amersham, Arlington Heights, Ill.).
  • ECL Plus chemiluminescence system Amersham, Arlington Heights, Ill.
  • SDS-PAGE gels for every test sample, were prepared in duplicate, containing the same amount of protein; and the control gel was stained with coomassie blue. The other gel was used for Western Blot analysis.
  • the intensity of the bands was analyzed by densitometry with a GS-800 Calibrated Imaging Densitometer (Quantity One 4.0.3 software; Bio-Rad). The results showed that AS-C significantly increased Fas expression of GBM cells (1 to 159 fold) but not Fas-L expression. In addition, the activation of the death receptor-induced apoptosis-related caspases-8 was monitored. The results indicated that the amount of procaspase-8 was only slightly increased at 6 h after AS-C treatment, whereas the amount of the activated caspase-8 was greatly increased at 6 h after AS-C treatment (see FIG. 3 a ).
  • procaspase-9 and procaspase-3 were also determined. Both procaspase-9 and procaspase-3 were highly activated at 6 h after AS-C treatment ( FIG. 3 c ).
  • BP increased p53, p21 and p16 expression by 1.4, 2.3 and 3.1 times respectively at 48 h. It also increased p53 phosphorylation by 5.2 times at 1.5 h and 9.2 times at 48 h, but decreased Rb phosphorylation by 0.2 times at 48 h ( FIG. 3 f ). Beta-actin was used as an internal control in this study. In FIG. 3 g, BP was seen to decrease also cdk2, cdk4, cdk6 cyclins D1 and cycline E.
  • the RG2 cells (rat GBM) and DBTRG-05MG cells (human GBM) were used in animal experiments to monitor the anti-tumor activities of AS-C and BP.
  • Male F344 rat (230-260 g) and male Foxn1 nu/nu mice (10-12 weeks) were obtained from National Laboratory Animal Center (Taipei, Taiwan). All procedures were performed in compliance with the Standard Operation Procedures of the Laboratory Animal Center of Tzu Chi University (Hualien, Taiwan). Animals were kept under pathogen-free conditions and fed a standard laboratory diet.
  • the DBTRG-05MG cells (human GBM) and RG2 cells (rat GBM) were prepared for nude mice xenografts and rat allogenics, respectively.
  • Syngeneic F344 rats in two groups (6/group) were implanted subcutaneously on the back with 1 ⁇ 10 6 RG2 cells.
  • the animals were administered by subcutaneous injection either with AS-C (500 mg/kg/day) (treatment group), or with the vehicle (50 mg/ml propylene glycol and 100 mg/ml Tween-80 in distilled water; Standard Chem. & Pharm., Tainan, Taiwan) (control group), at a spot distant from the inoculated tumor sites (>2 cm), on day 3, 6 and 9, after tumor cell implantation.
  • Tumor sizes were measured using a caliper and the volume was calculated as L ⁇ H ⁇ W ⁇ 0.52.
  • the animals were sacrificed when tumor volume exceeded 25 cm 3 and the day of sacrifice was assumed as the final survival day for the animals.
  • Experiment 2 Comparative effects of AS-C administered by subcutaneous injection and by intra-peritoneal injection on the tumor size of mice bearing subcutaneous human GBM tumor
  • Tumor sizes were measured using a caliper and the volume was calculated as L ⁇ H ⁇ W ⁇ 0.52.
  • the animals were sacrificed when tumor volume exceeded 1000 mm 3 in mice, and the day of sacrifice was assumed as the final survival day for the animals.
  • Experiment 3 Effect of AS-C administered by subcutaneous injection, on the tumor size of rats bearing in situ GBM tumor (intra-cranial allogenic GBM).
  • RG2 cells The cytotoxic effect of AS-C on in situ tumor was determined with RG2 cells.
  • Syngeneic rats in two groups (6/group) were implanted i.c. (striatum) with 5 ⁇ 104 RG2 cells, and treated with AS-C (500 mg/kg/day) or vehicle s.c. at day 4, 5, 6, 7 and 8 after tumor cell implantation.
  • Tumor volumes were measured and calculated by 3-T unit MRI (General Electric, Wis., USA) with echo-planar imaging capability (Signa LX 3.8, General Electric, Wis., USA) in Buddhist Tzu Chi General Hospital (Hualien, Taiwan). Briefly, rats were anesthetized with chloral hydrate (400 mg/ml, 1 ml/100 g).
  • Functional MRI scanning was conducted with a fast spin echo, echo-planar acquisition sequence in which the repetition time was 6000 msec, the echo time was 102 msec, the matrix image was 256 by 256, the field of view was 5 by 5 cm, and the in-plane resolution was 80 ⁇ m. Twenty slices, each 1.5 mm thick, were obtained every 19.5 seconds for 6.5 minutes for each rat.
  • the tumor was allowed to grow to a larger size to simulate a clinical condition where surgical removal of tumor was not an acceptable option.
  • DBTRG-05 MG cells (5 ⁇ 10 6 ) were implanted s.c. on the backs of nude mice.
  • the tumor-bearing mice were treated with single dose of AS-C (500 mg/kg) or vehicle (s.c.) only when the tumor volumes were ⁇ 250 mm 3 .
  • the mice were sacrificed to determine the cytotoxicities in tumors by H&E tissue staining at day 10 after treatment of AS-C. The tissue sections were observed and photographed under a light microscope at magnifications of 50 ⁇ and 400 ⁇ .
  • Experiment 1 Effects of BP on intracranial (i.c.) rat allogenics GBM tumors in F344 male rat.
  • the rats in two groups (6/group) were implanted i.c. (striatum) with 5 ⁇ 104 RG2 cells, and randomly treated with BP (300 mg/kg/day) or vehicle s.c. in the hind flank region after tumor cell implantation at day 4, 5, 6, 7 and 8 for five dosages.
  • Tumor volume was measured and calculated by MRI.
  • MRI was performed with a 3-T unit (General Electric, Wis., USA) with echo-planar imaging capability (Signa LX 3.8, General Electric, Wis., USA). Briefly, rats were anesthetized with chloral hydrate (400 mg/ml, 1 ml/100 g).
  • Functional MRI scanning was conducted with a fast spin echo, echo-planar acquisition sequence in which the repetition time was 6000 msec, the echo time was 102 msec, the matrix image was 256 by 256, the field of view was 5 by 5 cm, and the in-plane resolution was 80 ⁇ m. Twenty slices (1.5 mm thick each) were obtained every 19.5 seconds for 6.5 minutes for each rat. Finally, the whole tumor sizes (mm 3 ) were measured and calculated for each group.
  • FIG. 8 represents a mean ⁇ SE (*: p ⁇ 0.05; **: p ⁇ 0.001).
  • the mean of tumor volumes at day 14 and day 16 were 69.9 ⁇ 4.81 mm3 and 126.43 ⁇ 11.07 mm 3 for the control group respectively; and 46.6 ⁇ 1.8 mm 3 and 91.68 ⁇ 8.3 mm 3 for the BP-treated group respectively.
  • MRI image data showed that the tumor volume in situ of BP-treated group had a smaller region than control group.
  • Experiment 2 Effects of BP on supressing the growth of s.c. xenograft human GBM tumors in nude mice, and on the survival rate of the mice.
  • telomere activity was measured by the modified telomere repeat amplification protocol (TRAP) assay as described in the literature.
  • the pelleted cells were lysed with 200 ⁇ l of ice-cold lysis buffer (10 mM Tris-HCl, pH 7.5, 1 mM EGTA, 0.5% CHAPS, 10% [v/v] glycerol, 5 mM ⁇ -2-mercaptoethanol, and 0.1 mM phenylmethylsulfonyl fluoride) and incubated on ice for 30 minutes, and then centrifuged at 13,000 ⁇ g at 4° C. for 20 minutes. The supernatant extracts were quantified for protein using a BCA Protein Assay Kit (Pierce, Ill., USA).
  • TRAP assay was performed using a TRAPeze Telomerase Detection Kit (Intergen Co., Purchase, N.Y., USA) and the procedures were followed in accordance with the manufacturer's protocol.
  • a volume of the extract containing 0.5 ⁇ g of protein was added to 50 ⁇ l of reaction mixtures containing 0.1 ⁇ g of substrate oligonucleotide (TS) primer (5′-AATCCGTCGAGCAGAGTT-3′), 0.1 ⁇ g of TSK1 template (internal control), 0.1 ⁇ g of reverse oligonucleotide primer (RP), 2 U of Takara Taq DNA polymerase (Takara Shuzo Co., Japan), 20 mM Tris-HCl, pH 8.3, 1.5 mM MgCl2, 63 mM KCl, 0.005% (v/v) Tween 20, 1 mM EGTA, 50 ⁇ M each of deoxynucleotide triphosphate (dNTPs), and 1.25 ⁇
  • the reaction mixtures were incubated at 94° C. for 2.5 minutes and then amplified for 30 cycles of polymerase chain reaction (PCR) amplification at 94° C. for 30 seconds, 59° C for 30 seconds, and 72° C. for 90 seconds in a DNA thermal cycler (GeneAmp PCR System 2400, PerkinElmer Co., Norwalk, Conn., USA).
  • PCR polymerase chain reaction
  • the TRAP products were resolved by 12.5% (w/v) non-denaturing polyacrylamide gel electrophoresis (PAGE) in a buffer containing 54 mM Tris-HCl, pH 8.0, 54 mM boric acid, and 1.2 mM EDTA.
  • the gel was dried on filter paper for 1 hour and exposed on the X-film (Bio-Max MR, Kodak Rochester, N.Y., USA) at ⁇ 80° C. for 6 hours with an intensifying screen.
  • the signal intensity of the TRAP assay DNA ladder products was quantified by the Bio-Profil Biolight imaging analysis software, V2000.01 (Vulber Lourmat, France) and compared.

Abstract

The invention provides an acetone extract, chloroform extract or hexane extract of Angelicae sinensis and/or the active components purified therefrom, such as n-butylidenephthalide, which are effective in treating cancers.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 60/616,636, filed Oct. 8, 2004, the disclosure of which is hereby incorporated by reference herein.
  • BACKGROUND OF THE INVENTION
  • The invention mainly relates to a new use of an acetone extract, chloroform extract or hexane extract of Angelicae sinesis and the active components purified therefrom in the treatment of cancers.
  • Cancers are abnormal cell proliferations that result from the accumulation of genetic changes in cells endowed with proliferative potential. Treatment of cancers has relied mainly on surgery, chemotherapy, radiotherapy and more recently immunotherapy. However, new approaches for treating and preventing cancers are still desired.
  • Angelicae sinensis (Dangqui) is one of the most frequently occurring drugs in the prescriptions of traditional Chinese medicines. The traditional uses of Angelicae sinensis include those to promote blood production, protect liver, lower blood pressure, kill bacteria, ease pain mostly for menstrual disorder in women, and lower cholesterol (Chinese Herbs, Shanghai Science and Technology Publication, Inc., Shanghai, China, Vol. 5, p. 893, 1999).
  • CN1053747 disclosed that Angelicae sinensis (Oliv) Diels, ASD, and the ASDP and ASDE as effective components of an adjuvant was prepared, and could be used as an immunological adjuvant to genetically-engineered hepatitis B vaccines. It was reported in CN1109356 that the effective component, lactones (ASDE), extracted from Angelicae sinensis (oliv) diels, ASD, could be used as an immunological adjuvant, which can enhance the immunogenicity and help lower toxicity. Kumazawa et al. provided immunostimulating polysaccharides separated from a hot water extract of Angelicae sinensis, which could be used as a potent adjuvant for its anti-tumor activity as observed in the prolongation of the survival period of mice bearing Ehrlich ascites cells (Y. Kumazawa, et al., Immunology, Vol. 47, p. 75, 1982). However, this prior art reference provides only a general description of the treatment of cancers with the polysaccharides separated from Angelicae sinesis through their immunostimulating activity, without sufficient evidence regarding the mechanism.
  • BRIEF SUMMARY OF THE INVENTION
  • This invention provides that an acetone extract, chloroform extract or hexane extract of Angelicae sinensis, or at least one component purified therefrom, such as n-butylidenephthalide (BP), can also inhibit telomerase activity of cancer cells and further induce their apoptosis so that they can be used to treat malignant neoplasms. Therefore, an acetone extract, chloroform extract or hexane extract of Angelicae sinenses, and the components purified therefrom, such as n-butylidenephthalide, are potent for manufacturing of medicines for the treatment of cancers, and can be used in combination with chemotherapy drugs through their activities on cell cycle regulation, and telomerase inhibition.
  • Accordingly, one object of the present invention is to provide a method for inhibiting cancer cell proliferation and migration in tumor tissues.
  • Another object of the present invention is to provide a method for inhibiting telomerase activity of cancer cells.
  • Yet another object of the present invention is to provide a method for inducing apoptosis of cancer cells.
  • Another object of the present invention is to provide the use of an acetone extract, chloroform extract or hexane extract of Angelicae sinenses, or at least one component purified therefrom, such as n-butylidenephthalide, for manufacturing medicine for the treatment of cancer, and as an adjuvant in combination with chemotherapy drugs through their activities on cell cycle regulation and/or telomerase inhibition.
  • BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
  • The foregoing summary, as well as the following detailed description of the invention, will be better understood when read in conjunction with the appended drawings relating to embodiments which are presently preferred. It should be understood, however, that the invention is not limited to the precise embodiments shown.
  • In the drawings:
  • FIG. 1 a provides the results of the cell cycle analysis, which demonstrates that treatment with 70 μg/ml AS-C (the chloroform extract of Angelicae sinensis) enhanced cell cycle accumulation at G0/G1 phase (>90%) in GBM cells (DBTRG-05M) (*p<0.05) with a concurrent decrease in S phase. The results on G5T/VGH were about the same and not shown in the graph.
  • FIG. 1 b provides the results of the cell cycle analysis, which demonstrates that the treatment with 400 μM BP enhanced cell cycle accumulation at G0/G1 phase (>90%) in GBM cells (DBTRG-05MG) (*p<0.05, **p<0.005) with concurrent decrease in S phase.
  • FIG. 2 shows the effect of BP (n-butylidenephthalide) of 5 to 800 μM, in inducing GBM tumor cell (DBTRG-05MG) apoptosis, as assessed by TUNEL method, using propidium iodide as a counter staining (*p<0.05, ** p<0.005, *** p<0.0005).
  • FIGS. 3 a-3 c provide the results of the analyses of apoptosis pathways induced by AS-C, 70 μg/ml (wherein the DBTRG-05MG cell line was used.)
  • FIGS. 3 d-3 g provide the results of the analyses of apoptosis pathways induced by BP, 400 μM (wherein the DBTRG-05MG cell line was used.)
  • FIG. 4 shows the inhibitory effect of the AS-C treatment (500 mg/kg) on the tumor sizes in mice bearing subcutaneous GBM tumors (RG-2) (p<0.05).
  • FIG. 5 shows that the survival rate of the AS-C treated mice (dose—500 mg/kg) was significantly prolonged as compared with the control group (p<0.0001), wherein the DBTRG-05MG cell line was used.
  • FIG. 6 shows the inhibitory effect of AS-C treatment (500 mg/kg) on the growth of in situ GBM tumor (RG2) volume on rats (* p<0.05, ** p<0.001).
  • FIG. 7 shows the inhibitory effect of the AS-C treatment (500 mg/kg by intra-peritoneal or subcutaneous administration) on the xenograft tumor growth of mice (p<0.005), wherein the DBTRG-05MG cell line was used.
  • FIG. 8 shows the inhibitory effect of the BP treatment (300 mg/kg) on tumor volume of GBM in situ (RG2) on rats, which was calculated with MRI imaging using echo-planar imaging capability (* p<0.05, ** p<0.001).
  • FIG. 9 shows the inhibitory effect of BP treatment, at different dosages (70 to 800 mg/kg), on xenograft tumor growth on mice (p<0.005), wherein the DBTRG-05MG cell line was used.
  • FIG. 10 shows the effect of BP treatment (70 to 800 mg/kg) on the prolongation of survival period of nude mice with xenograft tumor (subcutaneous DBTRG-05MG) (p<0.001).
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention provides that the organic solvent extracts of Angelicae sinensis, or the components purified therefrom, such as n-butylidenephthalide (BP), can inhibit telomerase activity of cancer cells and further induce their apoptosis. Therefore, they can inhibit cancer cell proliferation and can be used to treat cancers.
  • Preparation of Angelicae sinensis Extracts
  • Angelicae sinensis (Dangqui) has long been used in blood diseases and female diseases. Normally, the dried root of Angelica sinensis (Oliv.) Diels, belonging to the family of Umbelliferae, is used. Angelica sinensis (AS) is appreciated by those skilled in this art. A variety of techniques are well known in the art for extracting, separating, and/or purifying individual active components of Angelicae sinensis. The organic solvent extracts of Angelicae sinensis may be obtained by any standard procedures commonly used in the field. According to the invention, Angelicae sinensis is extracted with acetone, chloroform, or hexanes. In one embodiment of the invention, the dried and powdered rhizomes of Angelicae sinensis were extracted with acetone as a solvent to give an extract as AS-A. Furthermore, AS-A was further extracted with chloroform to give an extract as AS-C; and AS-A was further extracted with hexanes to give an extract as AS-H.
  • Purification of Active Components
  • Active components of Angelicae sinensis may be isolated and/or purified from the organic solvent extracts of Angelicae sinensis by using any techniques known in the art. The active components may be purified from Angelicae sinensis in any form, particularly the rhizomes. Various techniques that may be employed in the further purification include filtration, selective precipitation, extraction with organic solvents, extraction with aqueous solvents, column chromatography, high performance liquid chromatography (HPLC), etc. According to the invention, some active components were purified from the organic solvent extracts of Angelicae sinensis, such as ligustilide and n-butylidenephthalide, which can induce tumor cell apoptosis. In one embodiment of the invention, E- and Z-geometrical isomers of n-butylidenephthalide (BP) were separated with column chromatography and characterized with HPLC and NMR.
  • Mechanisms of Cancer Treatment
  • Telomeres, the extremities of eukaryotic chromosomes, are essential for maintaining the integrity of the genome and are a key determinant of cellular aging and immortality (N. W. Kim, M. A. Piatyszek, K. R. Prowse, C. B. Harley, M. D. West, P. L. C. Ho, G. M. Coviello, W. E. Wright, S. L. Weinrich, J. W. Shay, Science, 266, 2011-2015 (1994)). Telomere length and the rate of its reduction vary among organs and individuals. Large interchromosomal variation in telomere length exists in mice and humans and an aberration of a telomere in a single chromosome can lead to abnormal chromosomal segregation (L. L. Sandell, V. A. Zakian, Cell, 75, 729-739 (1993)). Therefore, it is concluded that the regulation and maintenance of telomere length variation play an important role in cancer development. Apparently cells have a system to protect against both critical shortening and abnormal elongation of the telomere. Telomerase has been identified as one of the telomere length regulators (G. B. Morin, Cell, 59, 521-529 (1989)). Hence, any compound or substance having a selective inhibiting telomerase activity can inhibit tumor cell growth and thus further induce cell apoptosis of tumor cells.
  • Apoptosis is another mechanism of cancer therapy, which has become one of the newest areas of cell biology research.
  • The activation of the apoptosis program is regulated by various signals from both intracellular and extracellular stimuli. Indeed, in recent years evidence is beginning to accumulate that many (and perhaps all) agents of cancer chemotherapy kill tumour cells by launching the mechanisms of apoptosis. New drugs associated with apoptosis are expected to be most effective against tumours with high proliferation rates. Many such candidates are being screened for use in the treatment of cancer (Ricardo Pérez-Tomás, Beatriz Montaner, Esther Llagostera, Vanessa Soto-Cerrato, Biochemical Pharmacology, 66, 1447-1452 (2003)).
  • Apoptosis is monitored by the analysis of two commonly used endpoints—the morphological changes of cells (condensation of nuclear chromatin, formation of apoptotic bodies) and DNA fragmentation into large fragments (300 and 50 kbp) and then to oligonucleosomesized fragments (multiples of 200 bp), which appear as a “ladder” of DNA bands upon agarose gel electrophoresis. Although observation of these endpoints is an indicator of apoptosis, quantification of the percentage of apoptotic cells in a population by such an assay is impossible. For this purpose, we also used the TUNEL assay during which fluorescently-biotinylated nucleotides were added to the ends of DNA fragments within fixed cells (Jacques Piettea, Cédric Volantia, Annelies Vantieghemb, Jean-Yves Yvette Habrakena, Patrizia Agostinis, Biochemical Pharmacology, 66, 1651-1659 (2003)).
  • The relative contribution of the receptor and the mitochondrial pathways to drug-induced apoptosis has been a subject of controversy. It depends on the type of the cytotoxic drug itself, the dose and kinetcs or on differences between certain cell types, which affects the cell type dependent signaling in the Fas/FasL pathway.
  • Apoptosis pathways can be initiated through different sites, such as the plasma membrane, by way of death receptor mediated signaling (receptor pathway; Fas/FasL/caspase-8/caspase-3 pathway), the mitochondria (mitochondrial pathway; Bax/AIF/caspase-9/caspase-3 pathway), and cell cycle regulation (including p53, Rb tumor suppressors, p16 and p21 cyclin kinase inhibitors and cyclin/cdk cell cycle check points) (Simone Fulda, Matroulea, Klaus-Micheal Debatin, Cancer Letter, 197, 131-135 (2003)).
  • It has been reported in the literature that acetone extract, chloroform extract or hexane extract of Angelicae sinensis and its active components have anti-angina, anti-agglutination and certain other activities on the cardiovascular system.
  • Surprisingly, we found in this invention that the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, and the active components purified therefrom, such as n-butylidenephthalide, have anti-cancer activities.
  • According to this invention, the growth of several cancer cell lines were tested against the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, the active components purified therefrom, such as n-butylidenephthalide. It was found that they were cytotoxic to cancer cells; they inhibited telomerase activity of cancer cells (as shown in Example 7); they suppressed cancer cell proliferation (as shown in Example 2) and they also induced cancer cell apoptosis (as shown in Examples 3 and 4). Furthermore, animal studies also showed they were effective in suppressing cancer growth (as shown in Examples 5 and 6). They are therefore potent for treating cancers, particularly human malignant glioblastoma, colorectal cancer, leukemia, neuroblastoma, hepatoma, breast, ovarian and lung cancers.
  • Pharmaceutical Compositions
  • The acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives according to the present invention may be administered by any conventional route of administration including, but not limited to, oral, parenteral, intraperitoneal (ip), intravenous (iv), intramuscular (im), subcutaneous (sc), pulmonary, transdermal, buccal, nasal, sublingual, ocular, rectal, vaginal or other routes. It will be readily apparent to those skilled in the art that any dosage or frequency of administration that provides the desired therapeutic effect is suitable for use in the present invention. In a preferred embodiment of the invention, they are administered by oral delivery, using methods known to those skilled in the art of drug or food delivery.
  • For the purposes of therapeutic administration, the acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives may be in the form of a tablet, pill, capsule, granule, gel, powder, sterile parenteral solution or suspension, metered aerosol or liquid spray, or suppository, depending on the administration route. To prepare a pharmaceutical composition of the present invention, the organic solvent extracts of Angelicae sinensis, active components purified therefrom, or the derivatives are admixed with a pharmaceutically acceptable carrier according to conventional pharmaceutical compounding techniques, wherein the carrier may take a wide variety of forms depending on the form of preparation desired for administration. Suitable pharmaceutically acceptable carriers are well known in the art. Descriptions of some pharmaceutically acceptable carriers may be found in The Hand Book of Pharmaceutical Excipients, published by the American Pharmaceutical Association and the Pharmaceutical Society of Great Britain. For instance, the tablets, capsules, gels, solutions or suspensions may also include the following components: a pharmaceutically acceptable excipient or carrier, which is a non-toxic, inert solid or semi-solid, diluent, encapsulating material, a gel base or formulation auxiliary of any type. The solutions and suspensions may contain auxiliaries, such as water for injection, saline solution, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; proteins such as serum albumin to enhance solubility; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The solution or suspension preparations can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • The acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives according to the present invention may also be administered as an adjuvant in combination with chemotherapy drugs, such as actinomycin, adriamycin, Ara-C, bleomycin, carmustin, cisplatin, cyclophosphamide, daunomycin, mitomycin, taxol, vinblastine, etc.
  • The acetone extract, chloroform extract or hexane extract of Angelicae sinensis, active components purified therefrom, and the derivatives according to the present invention may also be formulated in any dietary compositions by using any techniques known to those skilled in the art.
  • The following Examples are given for the purpose of illustration only and are not intended to limit the scope of the present invention.
  • Materials and Methods
  • Preparation of Angelicae sinensis extracts and Compounds
  • The roots of Angelicae sinensis (Oliv.) were supplied from Chung-Yuan Co., Taipei, Taiwan and were identified by Professor Han-Ching Lin. A voucher of herbarium specimen was deposited at the School of Pharmacy, National Defense Medical Center. The dried and powdered rhizomes of Angelicae sinensis (12 kg) were extracted 3 times with acetone (24 L/time) to give an acetone extract called AS-A. AS-A was then subjected to chloroform extraction 3 times (24 L/times). The latter extracts were concentrated under reduced pressure to yield 31.67 g of chloroform extract, called AS-C (from 100 g of acetone extract). A hexane extract (AS-H) was obtained by extracting AS-A with hexanes. n-Butylidenephthalide (BP) was purchased from Lancaster Synthesis Ltd. (Newgate, Morecambe, UK) and used without further purification. E- and Z-forms of BP were separated with column chromatography and characterized with HPLC and NMR. They were dissolved in DMSO, incubated with shaking at 25° C. for 1 hour and stored at 4° C. before each in vitro experiment.
  • Cell Proliferation Assay in vitro
  • Human umbilical vascular endothelial cells (HUVECs) were purchased from Cascade Biologics, Inc. (USA). HUVECs were maintained in Medium 200 (Cascade Biologics, Inc. USA) supplemented with 10% fetal bovine serum (FBS; Gibco BRL) and low serum growth supplement (LSGS; Cascade Biologics, Inc. USA). Human dermal fibroblasts (HDFs) were purchased from Cascade Biologics, Inc. (USA). HDFs were maintained in Medium 106 (Cascade Biologics, Inc. USA) supplemented with 10% FBS and low serum growth supplement (LSGS; Cascade Biologics, Inc. USA). The human colon adenocarcinoma cell lines HT-29 were purchased from the ATCC (Manassas, Va. USA). HT-29 were maintained in Dubecco's Modified Eagle Medium (DMEM; Gibco) supplemented with 10% FBS and 100 ng/ml penicillin and 100 ng/ml streptomycin (Life Technologies, Inc., Grand Island, N.Y., USA). For the proliferation assay of cell in log growth (G1), the cells were harvested at 60-80% of confluence. 100 μl of cell suspensions were dispensed onto a Falcon 96-well plates The densities were 6×103 cells/well in Medium 200 (for HUVECs), and 9×103 cells/well in Medium 106 (for HDFs), and 5×103 cells/well in DMEM (for HT-29) supplemented with 10% FBS. The cells were pre-incubated for 24 hours in an incubator (Humidified atmosphere, e.g., at 37° C., 5% CO2). 10 μl of toxicant of various concentrations were added into the culture medium of the plate. The cell cultures were incubated for 48 hours. 10 μl of the Cell Counting Kit-8 (CCK-8; DOJINDO) solution were added and the cell cultures incubated for 1˜4 hours in the incubator. The absorbance was measured at 450 nm, using a microplate reader with a reference wavelength at 600 nm or over.
  • Extraction of RNA from HUVECs
  • Total RNA was isolated from the cultured stromal cells using a modified guanidium isothiocyanate method (Trizol; Invitrogen).
  • Homogenization: The cells were lysed directly in a 6 well culture plate by adding 1 ml of trizol reagent to a 3.5 cm diameter well, and passing the cell lysate several times through a pipette.
  • Phase separation: The homogenized samples were incubated for 5 minutes at 15 to 30° C. to permit the complete dissociation of nucleoprotein complexes. Then, 0.2 ml of chloroform (Riedel-de-haën) per 1 ml of trizol reagent was added. Sample tubes were capped securely. The tubes were vigorously shaken by hand for 15 seconds and incubated at 15 to 30° C. for 2 to 3 minutes. The samples were centrifuged at no more than 12,000×g for 15 minutes at 2 to 8° C. Following centrifugation, the mixture separated into a lower red, phenol-chloroform phase, an interphase, and a colorless upper aqueous phase. RNA remained exclusively in the aqueous phase.
  • RNA precipitation: The aqueous phase was transferred to a fresh tube. The RNA was precipitated from the aqueous phase by mixing with isopropyl alcohol (Fluka). 0.5 ml of isopropyl alcohol was added for every 1 ml of trizol reagent which was added for homogenization initially. The samples were incubated at 15 to 30° C. for 10 minutes and centrifuged at no more than 12,000×g for 10 minutes at 2 to 8° C.
  • RNA wash: The supernatant was removed. The RNA pellet was washed once with 75% ethanol, using at least 1 ml of 75% ethanol for every 1 ml of trizol reagent, which was added for the homogenization initially. The sample was mixed by vortexing and centrifuged at no more than 7,500×g for 5 minutes at 2 to 8° C.
  • Re-dissolving the RNA: The supernatant was removed, and then the RNA pellet was dried. The RNA was dissolved in RNase-free water, and incubated for 10 minutes at 55 to 60° C. The RNA can be stored at −70° C.
  • Cell Lines
  • Human tumor cell lines (MCF-7, CL1-5, HT-29, Caco-2), human umbilical vascular endothelial cells (HUVEC), and human dermal fibroblasts (HDF) were tested for sensitivity on Angelicae sinensis extracts, ligustilide, n-butylidenephthalide and its derivatives in vitro. The DBTRG-05MG, BCM, HL-60 and J5 cells were grown in RPMI-1640 medium containing 10% fetal calf serum and 100 ng/ml penicillin and 100 ng/ml streptomycin at 37° C. in a humidified atmosphere with 5% CO2. The G5T/VGH, RG2, N18, SVEC and Balb/3T3 cells were cultured in DMEM with 10% fetal calf serum and 100 ng/ml penicillin and 100 ng/ml streptomycin at 37° C. in 5% CO2. Mycoplasma infection of the culture cells was excluded by PCR screening methods before each experiment.
  • EXAMPLE 1 Analysis of Cell Cytotoxicity
  • The effects on cell viability after the treatments with different concentrations of the Angelicae sinensis extracts or the active components purified therefrom were evaluated by modified MTT assay in triplicate. Briefly, the cells (5×103) were incubated into 96-well plates containing 100 μl of a growth medium. The cells were permitted to adhere for 24 hours, then treated with 100 μl of the herbal extracts or the active components dissolved in the medium. The control contained DMSO of ≦0.02% (v/v). After 24, 48 and 72 h incubation, the drug-containing medium was replaced by 50 μl of fresh medium, and cells in each well were incubated in 50 μl of 400 μg/ml MTT for 6-8 h. The medium and MTT were removed later and 100 μl of DMSO was added to each well and to the control, to dissolve the soluble components. Absorbance at 550 nm of the solutions was measured with MRX Microtiter Plate Luminometer (DYNEX, USA). The absorbance of untreated cells was considered as 100%. To evaluate the effects of the extracts or the active components on cell growth rate of GBM cells, 5×103 exponentially growing cells were treated with different concentrations for 24, 48, or 72 h. The cytotoxicity of each test substance was determined as an IC50 value, which represents the drug concentration required to cause 50% inhibition. All experiments in this study were performed in triplicate.
    TABLE 1
    Cytotoxicity (IC50) of the different Angelicae sinensis organic solvent
    extracts, BP and its derivatives on different cells lines.
    Cytotoxicity, IC50
    Normal Cells Cancer cells
    HUVEC HDF Caco-2 MCF-7 RG2
    Extract AS-A 91.85 >100 >100 92.06 6.83
    (μg/ml) AS-C 44.19 85.08 51.17 >100 7.22
    AS-H >100 66.55 >100 62.42 30.49
    BP >100 300 >20 >20 1.4
    (>532 μM) (1596 μM) (>106 μM) (>106 μM)  (7.45 μM)
    BP-E form >100 >100 >20
    (>532 μM) (>532 μM) (>106 μM)
    BP-Z form >100 65.25 >20
    (>532 μM)  (347 μM) (>106 μM)

    HUVEC: Human umbilical vascular endothelial cells

    HDF: Human dermal fibroblasts

    Caco-2: Human colon adenocarcinoma

    MCF-7: Human breast carcinoma

    RG2: Rat malignant glioma

    SVEC: SV40 transformed mouse lymph node endothelial cell

    Balb/3T3: Mouse fibroblast cell
  • TABLE 2
    Cytotoxicity (IC50) of the organic solvent extracts
    of Angelicae sinensis, and BP on different cell lines.
    AS-A AS-C AS-H BP
    (μg/ml) (μg/ml) (μg/ml) (μg/ml)
    A549 90-110 8-12 (42.6-63.8 μM)
    AT12 100 10-15 (53.2-79.8 μM)
    J5 70-90  15-20 (79.8-106.4 μM)
    HCT15 80-100 25-30 (133.0-160.0 μM)
    HT-29 21-94  20 30 15-97 (79.8-516.0 μM)
    CL1-5 29 22 28 >100 (>532 μM)
    DBTRG-05MG 40-110 44 >400 7-10 (37.2-53.2 μM)
    G5T/VGH 50-223 46-60
    N18 111 35
    BCM 300 142 >400
    HL-60 367 173
    RG2 35 30 1.4 (7.45 μM)
    SVEC 76 86
    Balb/3T3 38 >400 >400 >300 (>1596 μM)

    A549, AT12: Human lung adenocarcinoma cell lines (AT12 and A549 are taxol-resistant subclones)

    J5: Human hepatoma cell line

    HCT15: Human colon adenocarcinoma cell line

    HT-29: Human colon adenocarcinoma cell line

    CL1-5: Human lung adenocarcinoma

    DBTRG-05MG: Human glioblastoma multiform cell line

    G5T/VGH: Human glioblastoma multiform cell line

    N18: Neuroblastoma

    BCM: Human breast carcinoma

    HL-60: Human promyelocytic leukemic cell

    RG2: Rat maglinant glioma

    SVEC: SV40 transformed mouse lymph node endothelial cell

    Balb/3T3: Mouse fibroblast cell
  • TABLE 3
    Test results of ETS-1, MMP-2, cell migration and
    tube formation of HUVEC
    Inhibition
    RT-PCR Tube
    ETS-1 MMP-2 Migration Formation
    Extract AS-A 1 1 30 30
    (μg/ml) AS-C 30 30 1 10
    AS-H 100 100 30 10
    BP 53.13 159.4 531.3
    (282.6 μM) (847.9 μM) (2826 μM)
    BP-E form 159.4
    BP-Z form 159.4 531.3 159.4
  • Both the n-butylidenephthalide (BP) and the Angelicae sinensis extracts (AS-A, AS-C, AS-H,) generally displayed a lower value of IC50 to a number of human tumor cell lines (Table 1 and Table 2) in comparison with the normal cells (HUVEC and HDF). BP, in particular, exhibited strong cytotoxic effect on human brain tumor cells. BP was also cytotoxic to two taxol-resistant human lung adenocarcinoma cells, a human hepatoma cell and two human colon adenocarcinoma cells.
  • The IC50 of AS-C and BP to brain tumor cell lines were 35˜60 μg/ml and 1.4˜10 μg/ml, respectively, while those to the normal cell line (HDF) were 85˜300 μg/ml, (p<0.0001).
  • Among the normal cells, the vascular endothelial cells (IC50=44.2±0.1 μg/ml) were more sensitive than fibroblast cells (IC50=85.1 μg/ml) to AS-C (p<0.05).
  • The inhibition effects of carmustin (BCNU) and Taxol were also tested and compared. The results showed that GBM tumors were not sensitive to carmustin (IC50>100 μg/ml) but DBTRG-05MG and G5T/VGH GBM cells were sensitive to Taxol (IC50=61.0±3.3 μg/ml and IC50<0.1 μg/ml, respectively). However, Taxol induced a very high cytotocixity (IC50<0.1 μg/ml), which is much greater than that induced by AS-C and BP, in vascular endothelia cells. After treatment with AS-C or BP, the GBM cells (DBTRG-05MG) were seen to be detached and floating in the media at different points of time within a 72-hour period of observation. The extent of GBM cell detachment and flotation was found to increase with time, and with the increase in dosage (in the case of BP when an observation was made at 3 hours).
  • The GBM cell detachment and flotation after treatment with AS-C or BP, can be attributed to morphology change in the tumor cells. In the above experiment, BCNU (carmustin) was used as the system control.
  • EXAMPLE 2 AS-C and BP Enhance the Cell Cycle Arrest at G0/G1 Phase in GBM Cells
  • Brain tumor cell lines DBTRG-05MG and G5T/VGH were cultured in the growth medium with a diluent. For each test and control, DMSO was added, and the content is less than 0.02% (v/v). For the AS-C and BP treatment, 70 μg/ml of AS-C and 400 μM of BP were added, respectively. All were cultured for 48 hours. The analysis of cell cycle distribution was performed by DNA staining with propidium iodide (PI). Briefly, 2×106 adherent cells were detached by trypsinization. The detached cells and the floating dead cells were centrifuged and washed twice with 10 ml of cold 1×PBS (Life Technologies, Inc.). Supernatant was aspirated, cells were re-suspended in 0.8 ml of 1×PBS, and then 200 μl of PI solution (50 μg/ml PI+0.05 mg/ml RNase A; Sigma Chemical Co.) was added, and the cells were refrigerated at 4° C. overnight. The cells were incubated while protected from light at room temperature for at least 2 h before DNA analysis. After staining, DNA was detected and quantified on 20,000 total cells using a FACScan (Becton Dickinson Immunocytometry Systems, San Jose, Calif., USA) and CellQuest analysis software. The G0/G1 phases were gated in M1 (×2); G2/M phases were gated in M2 (×2); the total cells were gated in M3; S phase was M3−(M1(×2)+M2(×2)); Sub G1 phase (apoptosis cells) was gated in M4.
  • The cell cycle analysis demonstrated that both 70 μg/ml of AS-C and 400 μM of BP enhanced the cell cycle accumulation at G0/G1 phase (>90%) in GBM cells. FIGS. 1 a and 1 b showed that both AS-C and BP enhanced a significant G0/G1 phase arrest with a concurrent decrease of S phase after treatment for 12 hours to 48 hours (p<0.05,p<0.005).
  • EXAMPLE 3 AS-C and BP Induce GBM Cells Apoptosis
  • Apoptotic cell death was analyzed using In Situ Cell Death Detection Kit, POD (Roche, Germany). Changes in DNA chromatin morphologic features were used for quantification. The procedures were performed in accordance with the manufacturer's instructions. Briefly, cells were cultured on culture dish and analyzed 72 hours after treatment with AS-C (70 μg/ml) and BP (5˜800 μg/ml), respectively. In AS-C and BP-treated groups, the suspended cells were collected. In the control group, adherent and floating cells were collected. Then, the cells were fixed with 3.7% formaldehyde at room temperature for 15 min. on saline coated slides, washed once in 1×PBS, and incubated in cold permeabilization solution (0.1% Triton X-100+0.1% sodium citrate) after reducing activity of endogenous peroxidase with 3% H2O2. The cells were washed with 1×PBS again, and incubated with terminal deoxynucleiotidyl transferase (TdT)-mediated dUTP nicks labeling (TUNEL) reaction mixture for 60 minutes at 37° C. Then, the cells were washed with 1×PBS, counterstained with propidium iodide (PI) for cell counting. For quantification of apoptosis, the results were viewed under fluorescence microscopy (Nikon, Kawasaki, Japan).
  • When compared to untreated cells, almost all detached GBM cells in the AS-C and BP-treated groups were found to have undergone apoptosis. The apoptotic cells with AS-C treatment were detected by fluorescence microscopy (400×), with In Situ TUNEL staining and propidium iodide cell counterstaining, and observed under light field. Likewise, BP induced apoptosis in GBM cells was assessed by TUNEL method and using propidium iodide as a counter stain. The GBM cells were exposed to BP (5 to 800 μM) for 48 hours before assessment. The results were shown in FIG. 2. It was found that as compared to the untreated cells (control), the apoptosis rates of GBM cells in the BP treatment group, were much higher.
  • EXAMPLE 4 AS-C and BP Induce Apoptosis through the Activation of Multiple Pathways
  • Western blot analysis of apoptosis molecules
  • DBTRG-05MG cells (human GBM cells) were treated with AS-C (70 μg/ml) for 0, 6, 12, 24 and 48 hours. In another test, DBTRG-05MG cells were treated with BP (400 μM) for 0, 1.5, 3, 6, 12, 24 and 48 hours. The cell pellets were re-suspended in lysis buffer (10 nM Tris-HCl, pH 7.5, 1 mM EGTA, 0.5% CHAPS, 10% (v/v) glycerol, 5 mM , β-2-mercaptoethanol and 0.1 mM phenylmethylsulfonyl fluoride) and incubated on ice for 30 min, and then centrifuged at 13000×rpm at 4° C. for 20 minutes. The protein concentration of whole cell lysates was measured with BCA protein assay kit (Pierce, Rockford, Ill.) following the manufacturer's instructions. The cell lysates (20 μg/lane) were electrophoresed on 10-12% SDS-PAGE (Bio-Rad, Hercules, Calif.). Proteins were transferred to polyvinyldenefluoride (PVDF) membranes (Amersham Lifesciences, Piscataway, N.J.). The membranes were masked for 1 hour at room temperature with 5% skim milk as the blocking agent, and incubated with the respective antibodies of Fas (FL-335), Fas-L (C-178), caspase 3 (H-277), caspase 8 (H-134), caspase 9 (H-170), Bax (B-9), p16 (F-12), p21 (F-5), p53 (DO-1; 1/100 dilution) (Santa Cruz Biotechnology Inc., Calif., U.S.A.), phospho-p53 (Ser15; 1/2000 dilution) and phospho-Rb (Ser795; 1/2000 dilution) (Cell Signaling Technology, Mass., USA) for 2 hours at room temperature. Antibody recognition was detected, by incubating the membranes with the respective anti-mouse, anti-rabbit, anti-goat IgG secondary antibodies ( 1/1000 dilution; Santa Cruz Biotechnology Inc., Calif., U.S.A.) conjugated to horseradish peroxidase, for 1 hour at room temperature, and visualized with the ECL Plus chemiluminescence system (Amersham, Arlington Heights, Ill.). For system control, SDS-PAGE gels, for every test sample, were prepared in duplicate, containing the same amount of protein; and the control gel was stained with coomassie blue. The other gel was used for Western Blot analysis. The intensity of the bands was analyzed by densitometry with a GS-800 Calibrated Imaging Densitometer (Quantity One 4.0.3 software; Bio-Rad). The results showed that AS-C significantly increased Fas expression of GBM cells (1 to 159 fold) but not Fas-L expression. In addition, the activation of the death receptor-induced apoptosis-related caspases-8 was monitored. The results indicated that the amount of procaspase-8 was only slightly increased at 6 h after AS-C treatment, whereas the amount of the activated caspase-8 was greatly increased at 6 h after AS-C treatment (see FIG. 3 a).
  • The phosphorylations of p53 and Rb proteins were monitored and the results showed the AS-C increased phosphorylated p53 protein at 6 h after treatment. Furthermore, the amount of total p53 protein was increased as well at 6 h and then gradually decreased. However, phosphorylated Rb protein was seen to decrease at 6 h, and became undetectable at 12 h after AS-C treatment. These results indicated that AS-C could trigger the cell cycle checkpoint machinery. The amounts of p16, p21 and Bax in AS-C treated GBM cells were consequently measured and all of these three proteins were found to increase after treatment with AS-C (see FIG. 3 b).
  • Finally, the activations of procaspase-9 and procaspase-3 were also determined. Both procaspase-9 and procaspase-3 were highly activated at 6 h after AS-C treatment (FIG. 3 c).
  • In the case of BP, the results showed that BP 400 μM greatly increased the expression of Fas (from 5.2 times at 1.5 h to 27.9 times at 48 h), while suppressing the expression of the Fas Ligand on the GBM cells (see FIG. 3 d).
  • It is also observed that BP enhanced the activation of caspase 8, which was increased to 137.9 by the 48 h, while procaspase 8 declined (see FIG. 3 d).
  • The study of the role of mitochondrial pathway in BP-induced apoptosis showed that BP induced Bax and AIF expression, which increased to 16 times and 2.4 times respectively, by the 48 h, and activated caspase-9 by 25.8 times at the 48 h while procaspase-9 declined (see FIG. 3 e). Caspase-3 was also observed to increase while procaspase-3 declined (FIG. 3 d).
  • The study of the role of cell cycle pathway in BP-induced apoptosis showed that BP increased p53, p21 and p16 expression by 1.4, 2.3 and 3.1 times respectively at 48 h. It also increased p53 phosphorylation by 5.2 times at 1.5 h and 9.2 times at 48 h, but decreased Rb phosphorylation by 0.2 times at 48 h (FIG. 3 f). Beta-actin was used as an internal control in this study. In FIG. 3 g, BP was seen to decrease also cdk2, cdk4, cdk6 cyclins D1 and cycline E.
  • In conclusion, we theorize a schematic model of the apoptosis signal transduction pathways induced by BP stress, which consists of death receptor, mitochondrial and cell cycle pathways.
  • EXAMPLE 5 Animal Studies
  • The RG2 cells (rat GBM) and DBTRG-05MG cells (human GBM) were used in animal experiments to monitor the anti-tumor activities of AS-C and BP. Male F344 rat (230-260 g) and male Foxn1 nu/nu mice (10-12 weeks) were obtained from National Laboratory Animal Center (Taipei, Taiwan). All procedures were performed in compliance with the Standard Operation Procedures of the Laboratory Animal Center of Tzu Chi University (Hualien, Taiwan). Animals were kept under pathogen-free conditions and fed a standard laboratory diet. The DBTRG-05MG cells (human GBM) and RG2 cells (rat GBM) were prepared for nude mice xenografts and rat allogenics, respectively.
  • For AS-C treated group
  • Experiment 1—Effect of AS-C administered by subcutaneous injection on the survival rate and tumor size of rats bearing subcutaneous GBM tumor
  • Syngeneic F344 rats in two groups (6/group) were implanted subcutaneously on the back with 1×106 RG2 cells. The animals were administered by subcutaneous injection either with AS-C (500 mg/kg/day) (treatment group), or with the vehicle (50 mg/ml propylene glycol and 100 mg/ml Tween-80 in distilled water; Standard Chem. & Pharm., Tainan, Taiwan) (control group), at a spot distant from the inoculated tumor sites (>2 cm), on day 3, 6 and 9, after tumor cell implantation. Tumor sizes were measured using a caliper and the volume was calculated as L×H×W×0.52. The animals were sacrificed when tumor volume exceeded 25 cm3 and the day of sacrifice was assumed as the final survival day for the animals.
  • The results showed that AS-C treatment had a significant inhibitory effect on tumor growth when compared with the untreated (control) group (p<0.05) (FIG. 4). The average tumor size at day 26 was 20.7±1.5 cm3 for the control group and 11.5±0.7 cm3 for the treatment group, respectively. Survival of rats in the AS-C treated group was significantly prolonged, compared with those in the control group (40±2.7 days vs 30±2.1; p<0.0001) (FIG. 5).
  • With a dose of 500 mg/kg subcutaneous injection of AS-C, no drug related toxicities were observed in the rats as evidenced by the results of the body weights and histological analysis of the vital organs.
  • Experiment 2—Comparative effects of AS-C administered by subcutaneous injection and by intra-peritoneal injection on the tumor size of mice bearing subcutaneous human GBM tumor
  • Nude mice, in two groups (6 /group), were implanted s.c. with 5×106 DBTRG-05MG cells, and administered with AS-C (i.p. 500 mg/kg/day), AS-C (s.c. 500 mg/kg/day) or vehicle (s.c.) on day 5 after tumor cell implantation. Tumor sizes were measured using a caliper and the volume was calculated as L×H×W×0.52. The animals were sacrificed when tumor volume exceeded 1000 mm3 in mice, and the day of sacrifice was assumed as the final survival day for the animals.
  • The results showed that there were significant suppressions of tumor growth in the AS-C i.p. (500 mg/kg) and AS-C s.c. (500 mg/kg)-treated groups compared with the untreated group (p<0.005). The mean values of tumor sizes at day 38 were 849.9±150.1 mm3 in the control group, 295.5±25.3 mm3 in AS-C i.p. (500 mg/kg) treated group and 155.1±56.4 mm3 in AS-C s.c. (500 mg/kg) treated group. The results were shown in FIG. 7.
  • Experiment 3—Effect of AS-C administered by subcutaneous injection, on the tumor size of rats bearing in situ GBM tumor (intra-cranial allogenic GBM).
  • The cytotoxic effect of AS-C on in situ tumor was determined with RG2 cells. Syngeneic rats in two groups (6/group), were implanted i.c. (striatum) with 5×104 RG2 cells, and treated with AS-C (500 mg/kg/day) or vehicle s.c. at day 4, 5, 6, 7 and 8 after tumor cell implantation. Tumor volumes were measured and calculated by 3-T unit MRI (General Electric, Wis., USA) with echo-planar imaging capability (Signa LX 3.8, General Electric, Wis., USA) in Buddhist Tzu Chi General Hospital (Hualien, Taiwan). Briefly, rats were anesthetized with chloral hydrate (400 mg/ml, 1 ml/100 g). Functional MRI scanning was conducted with a fast spin echo, echo-planar acquisition sequence in which the repetition time was 6000 msec, the echo time was 102 msec, the matrix image was 256 by 256, the field of view was 5 by 5 cm, and the in-plane resolution was 80 μm. Twenty slices, each 1.5 mm thick, were obtained every 19.5 seconds for 6.5 minutes for each rat.
  • Significant declines of tumor volume in the treated group were observed in the MRI image data, compared with the untreated group (p<0.05) (FIG. 6). The mean tumor volumes at day 14 and day 16 were 70±4.8 mm3 and 126.4±11.1 mm3 in the control group versus 46.2±3.6 mm3 and 99.5±9.5 mm3 in AS-C treated group.
  • Experiment 4—Cytotoxicities of AS-C administered by subcutaneous injection, on the tumor size of mice bearing xenograft human GBM tumor
  • In this experiment, the tumor was allowed to grow to a larger size to simulate a clinical condition where surgical removal of tumor was not an acceptable option.
  • As described above, DBTRG-05 MG cells (5×106) were implanted s.c. on the backs of nude mice. The tumor-bearing mice were treated with single dose of AS-C (500 mg/kg) or vehicle (s.c.) only when the tumor volumes were ≧250 mm3. The mice were sacrificed to determine the cytotoxicities in tumors by H&E tissue staining at day 10 after treatment of AS-C. The tissue sections were observed and photographed under a light microscope at magnifications of 50× and 400×.
  • The photographs of histology analysis showed that AS-C had induced a nucleic degradation, a cavity cytosol and tumor cell death in the tumor cell mass. In contrast, the control tumor were growing very well and the cytotoxic effects as seen in the AS-C treated group, were not found in the tumor mass.
  • For BP treated group
  • Experiment 1—Effects of BP on intracranial (i.c.) rat allogenics GBM tumors in F344 male rat.
  • The rats in two groups (6/group) were implanted i.c. (striatum) with 5×104 RG2 cells, and randomly treated with BP (300 mg/kg/day) or vehicle s.c. in the hind flank region after tumor cell implantation at day 4, 5, 6, 7 and 8 for five dosages. Tumor volume was measured and calculated by MRI. MRI was performed with a 3-T unit (General Electric, Wis., USA) with echo-planar imaging capability (Signa LX 3.8, General Electric, Wis., USA). Briefly, rats were anesthetized with chloral hydrate (400 mg/ml, 1 ml/100 g). Functional MRI scanning was conducted with a fast spin echo, echo-planar acquisition sequence in which the repetition time was 6000 msec, the echo time was 102 msec, the matrix image was 256 by 256, the field of view was 5 by 5 cm, and the in-plane resolution was 80 μm. Twenty slices (1.5 mm thick each) were obtained every 19.5 seconds for 6.5 minutes for each rat. Finally, the whole tumor sizes (mm3) were measured and calculated for each group.
  • The results indicated that there was a significant reduction of tumor volume in the BP treated group as compared with the untreated group (p<0.05), as calculated using MRI scanning and Echo-planar as described above (FIG. 8). Each column in FIG. 8 represents a mean±SE (*: p<0.05; **: p<0.001). The mean of tumor volumes at day 14 and day 16 were 69.9±4.81 mm3 and 126.43±11.07 mm3 for the control group respectively; and 46.6±1.8 mm3 and 91.68±8.3 mm3 for the BP-treated group respectively. MRI image data showed that the tumor volume in situ of BP-treated group had a smaller region than control group.
  • Experiment 2: Effects of BP on supressing the growth of s.c. xenograft human GBM tumors in nude mice, and on the survival rate of the mice.
  • Animals (Foxn1 nude mice) in six groups (6/group) were implanted subcutaneously (s.c.) with 1×106 DBTRG-05MG cells, and randomly treated with BP s.c. (70, 150, 300, 500, 800 mg/kg/day) or vehicle s.c. at a site remote (>2 cm) from the incubated tumor after tumor cell implantation, at day 4, 5, 6, 7 and 8 for all five dosages. Tumor size was measured every 2 days and tumor volume was calculated. Animals were sacrificed when tumor exceeded 1000 mm3. Tumor growth was monitored for 3 months for those not sacrificed. Survival rate was followed for up to 200 days.
  • The results showed that there were significant suppressions of tumor growth in the BP-treated groups compared with the untreated group (FIG. 9; p<0.005 for the 300 mg/kg group), and the degree of tumor growth inhibition is dose dependent.
  • Log-rank (Mantel-Cox) comparison of survival plots given in FIG. 10 indicated that the survival period of nude mice with xenograft subcutaneous human GBM was prolonged up to 200 days upon treatment with BP.
  • EXAMPLE 7 Effects of AS-C on Telomerase Activity of Human Malignant Tumor Cell
  • Assay for telomerase activity by TRAP
  • The telomerase activity was measured by the modified telomere repeat amplification protocol (TRAP) assay as described in the literature. The pelleted cells were lysed with 200 μl of ice-cold lysis buffer (10 mM Tris-HCl, pH 7.5, 1 mM EGTA, 0.5% CHAPS, 10% [v/v] glycerol, 5 mM β-2-mercaptoethanol, and 0.1 mM phenylmethylsulfonyl fluoride) and incubated on ice for 30 minutes, and then centrifuged at 13,000×g at 4° C. for 20 minutes. The supernatant extracts were quantified for protein using a BCA Protein Assay Kit (Pierce, Ill., USA). TRAP assay was performed using a TRAPeze Telomerase Detection Kit (Intergen Co., Purchase, N.Y., USA) and the procedures were followed in accordance with the manufacturer's protocol. In brief, a volume of the extract containing 0.5 μg of protein was added to 50 μl of reaction mixtures containing 0.1 μg of substrate oligonucleotide (TS) primer (5′-AATCCGTCGAGCAGAGTT-3′), 0.1 μg of TSK1 template (internal control), 0.1 μg of reverse oligonucleotide primer (RP), 2 U of Takara Taq DNA polymerase (Takara Shuzo Co., Japan), 20 mM Tris-HCl, pH 8.3, 1.5 mM MgCl2, 63 mM KCl, 0.005% (v/v) Tween 20, 1 mM EGTA, 50 μM each of deoxynucleotide triphosphate (dNTPs), and 1.25 μCi of (γ32P) ATP, 3000 Ci/mmol (PerkinElmer Life Science, Boston, Mass., USA). The reaction mixtures were incubated at 94° C. for 2.5 minutes and then amplified for 30 cycles of polymerase chain reaction (PCR) amplification at 94° C. for 30 seconds, 59° C for 30 seconds, and 72° C. for 90 seconds in a DNA thermal cycler (GeneAmp PCR System 2400, PerkinElmer Co., Norwalk, Conn., USA). The TRAP products were resolved by 12.5% (w/v) non-denaturing polyacrylamide gel electrophoresis (PAGE) in a buffer containing 54 mM Tris-HCl, pH 8.0, 54 mM boric acid, and 1.2 mM EDTA. The gel was dried on filter paper for 1 hour and exposed on the X-film (Bio-Max MR, Kodak Rochester, N.Y., USA) at −80° C. for 6 hours with an intensifying screen. The signal intensity of the TRAP assay DNA ladder products was quantified by the Bio-Profil Biolight imaging analysis software, V2000.01 (Vulber Lourmat, France) and compared.
  • Statistics
  • Data were expressed as the mean±SD or SE. Statistical significance was analyzed by Student's t-test. A p value of <0.05 was considered significant. Survival was compared by log-rank (Mantel-Cox) test. Median survival time was estimated from Kaplan-Meier analysis.
  • While embodiments of the present invention have been illustrated and described, various modifications and improvements can be made by persons skilled in the art. It is intended that the present invention is not limited to the particular forms as illustrated, and that all the modifications not departing from the spirit and scope of the present invention are within the scope as defined in the appended claims.

Claims (14)

1. A method for inhibiting cancer cell proliferation and migration in tumor tissues in a subject in need thereof, comprising administering to the subject an acetone extract, chloroform extract or hexane extract of Angelicae sinenses or at least one active component purified from any of the Angelicae sinenses extracts in an amount effective for inhibiting cancer cell proliferation in tumor tissues.
2. A method for inhibiting telomerase activity of cancer cells in a subject in need thereof, comprising administering to the subject an acetone extract, chloroform extract or hexane extract of Angelicae sinenses or at least one active component purified from any of the Angelicae sinenses extracts in an amount effective for inhibiting telomerase activity of cancer cells.
3. A method for inducing apoptosis of cancer cells in a subject in need thereof, comprising administering to the subject an acetone extract, chloroform extract or hexane extract of Angelicae sinenses or at least one active component purified from any of the Angelicae sinenses extracts in an amount effective for inducing apoptosis of cancer cells.
4. A method for treating cancer in a subject in need thereof, comprising administering to the subject a medicine comprising an acetone extract, chloroform extract or hexane extract of Angelicae sinenses or at least one active component purified from any of the Angelicae sinenses extracts in an amount effective for at least one of preventing and treating the cancer.
5. A method for the treatment of cancer in a subject in need thereof, comprising administering to the subject an acetone extract, chloroform extract or hexane extract of Angelicae sinenses or at least one active component purified from any of the Angelicae sinenses extracts as an adjuvant in combination with one or more chemotherapy drugs through their activities on cell cycle regulation or telomerase inhibition.
6. The method according to any one of claims 1 to 5, wherein the active component purified from any of the Angelicae sinensis extracts includes n-butylidenephthalide.
7. The method according to any one of claims 1 to 5, wherein the cancer is selected from the group consisting of human malignant glioblastoma, colorectal, leukemia, neuroblastoma, hepatoma, breast, ovarian and lung cancer.
8. The method according to any one of claims 1 to 5, wherein the Angelicae sinenses extract or the at least one active component purified from any of the Angelicae sinenses extracts is administered via an oral, parenteral, intravenous (iv), intraperitoneal (ip), intravenous (iv), intramuscular (im), subcutaneous (sc), pulmonary, transdermal, buccal, nasal, sublingual, ocular, rectal or vaginal route.
9. The method according to claim 8, wherein the Angelicae sinenses extract or the at least one active component purified from any of the Angelicae sinenses extract is administered via an oral, parenteral, or intravenous route.
10. A method for the treatment of cancer in a subject in need thereof, comprising administering to the subject n-butylidenephthalide in an amount effective for treatment of the cancer.
11. A method for the treatment of cancer in a subject in need thereof, comprising administering to the subject n-butylidenephthalide in an adjuvant in combination with one or more chemotherapy drugs through their activities on cell cycle regulation or telomerase inhibition.
12. The method according to claim 10 or 11, wherein the cancers are selected from the group consisting of human malignant glioblastoma, colorectal, leukemia, neuroblastoma, hepatoma, breast, ovarian and lung cancer.
13. The method according to claim 10 or 11, wherein n-butylidenephthalide is administered via an oral, parenteral, intravenous (iv). intraperitoneal (ip), intravenous (iv), intramuscular (im), subcutaneous (sc), pulmonary, transdermal, buccal, nasal, sublingual, ocular, rectal or vaginal route.
14. The method according to claim 13, wherein n-butylidenephthalide is administered via an oral, parenteral, or intravenous route.
US11/246,009 2004-10-08 2005-10-07 Angelicae sinensis extracts useful for treatment of cancers Abandoned US20060110469A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US11/246,009 US20060110469A1 (en) 2004-10-08 2005-10-07 Angelicae sinensis extracts useful for treatment of cancers
US11/561,713 US7455861B2 (en) 2004-10-08 2006-11-20 Angelicae sinensis extracts useful for treatment of cancers
US12/260,723 US20090111870A1 (en) 2004-10-08 2008-10-29 Angelicae Sinensis Extracts Useful for Treatment of Cancers

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61663604P 2004-10-08 2004-10-08
US11/246,009 US20060110469A1 (en) 2004-10-08 2005-10-07 Angelicae sinensis extracts useful for treatment of cancers

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/561,713 Division US7455861B2 (en) 2004-10-08 2006-11-20 Angelicae sinensis extracts useful for treatment of cancers

Publications (1)

Publication Number Publication Date
US20060110469A1 true US20060110469A1 (en) 2006-05-25

Family

ID=35613784

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/246,009 Abandoned US20060110469A1 (en) 2004-10-08 2005-10-07 Angelicae sinensis extracts useful for treatment of cancers
US11/561,713 Active US7455861B2 (en) 2004-10-08 2006-11-20 Angelicae sinensis extracts useful for treatment of cancers
US12/260,723 Abandoned US20090111870A1 (en) 2004-10-08 2008-10-29 Angelicae Sinensis Extracts Useful for Treatment of Cancers

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/561,713 Active US7455861B2 (en) 2004-10-08 2006-11-20 Angelicae sinensis extracts useful for treatment of cancers
US12/260,723 Abandoned US20090111870A1 (en) 2004-10-08 2008-10-29 Angelicae Sinensis Extracts Useful for Treatment of Cancers

Country Status (7)

Country Link
US (3) US20060110469A1 (en)
EP (1) EP1645280B1 (en)
JP (1) JP4781771B2 (en)
AT (1) ATE455550T1 (en)
DE (1) DE602005019010D1 (en)
SG (1) SG121980A1 (en)
TW (1) TWI298259B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050191242A1 (en) * 2003-11-25 2005-09-01 Janice Brissette Foxn1 and pigmentation
US20110076303A1 (en) * 2009-06-25 2011-03-31 Tokuro Iwabuchi Methods for Screening for Anti-Graying Agents on the Basis of AFF-4
US20110165201A1 (en) * 2010-01-05 2011-07-07 National Dong Hwa University Anticancer formulation
US20140212970A1 (en) * 2013-01-25 2014-07-31 China Medical University Method and kit for culturing stem cells
US8927601B2 (en) * 2011-12-20 2015-01-06 National Dong Hwa University Uses of N-butylidenephthalide in treating a liver injury and improving liver function
US9504751B2 (en) 2014-04-24 2016-11-29 Everfront Biotech Inc. Stable pharmaceutical composition
EP3338775A4 (en) * 2015-08-19 2019-04-03 Everfront Biotech Inc. Use of butylidenephthalide
US10688128B2 (en) * 2016-05-23 2020-06-23 Everfront Biotech Inc. Use of Z-butylidenephthalide in activating autoimmune system
US10987339B2 (en) 2015-08-19 2021-04-27 Everfront Biotech Inc. Uses of butylidenephthalide
CN114989891A (en) * 2022-05-25 2022-09-02 湖南诺泽生物科技有限公司 Angelica essential oil and preparation method and application thereof

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005336866B2 (en) 2005-09-30 2012-06-14 Suzhou Herui Biomed Co., Ltd The use of phthalide derivatives
JP5161239B2 (en) * 2007-01-19 2013-03-13 チェン、フェイ Antitumor activity of dimers of phthalide compounds
CN101347474B (en) * 2007-07-20 2011-11-30 天津天士力制药股份有限公司 Effective component of Chinese angelica and preparation and use thereof
TWI386203B (en) * 2011-01-07 2013-02-21 Univ China Medical Pharmaceutical composition for treating brain cancer or reducing temozolomide-resistance of brain cancer cells and uses of the same
TWI486165B (en) 2011-02-15 2015-06-01 Univ China Medical Pharmaceutical compositions and extracts for inhibiting blood vessel stenosis and uses of the same
TWI460168B (en) * 2012-08-10 2014-11-11 Univ China Medical Pharmaceutical composition for inhibiting autophagy of motor neurons and uses of the same
TWI547280B (en) * 2014-04-24 2016-09-01 長弘生物科技股份有限公司 Stable pharmaceutical composition
TWI522099B (en) 2014-06-04 2016-02-21 中國醫藥大學 Pharmaceutical formulation for treating pancreatic cancer and uses of the same
KR101759637B1 (en) * 2014-09-26 2017-07-20 연천군 Health functional food for preventing liver cancer
CN106466315B (en) * 2015-08-19 2021-04-06 长弘生物科技股份有限公司 Use of butylene phthalide
CN109568308A (en) * 2017-12-28 2019-04-05 西南大学 Ligustilide is preparing the application in the drug for preventing and treating acute myeloid leukaemia
KR102350039B1 (en) * 2019-08-28 2022-01-11 경희대학교 산학협력단 Pharmaceutical composition for preventing or treating cancer comprising Angelica gigas, Aconitum carmichaeli Debeaux and Zingiber officinale Roscoe

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4613591A (en) * 1982-08-23 1986-09-23 Tsumura Juntendo Inc. Adminiculum for antitumor agents
US20030165580A1 (en) * 2002-03-04 2003-09-04 Xinxian Zhao Safe pharmaceutical composition for treatment and prevention of gynecological disease

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0352820A (en) * 1989-07-19 1991-03-07 Nagakura Seiyaku Kk Carcinogenic promotion inhibitor
CN1053747A (en) 1991-03-18 1991-08-14 中国医学科学院医药生物技术研究所 Preparation technology of vaccine immunity adjuvant for hepatitis b gene engineering
CN1057915C (en) 1994-09-19 2000-11-01 中国医学科学院医药生物技术研究所 Immunologic adjuvant
CN1058893C (en) * 1997-11-13 2000-11-29 王汝辉 Medicine for treating woman's dysmenorrhea and preparing process thereof
ATE319462T1 (en) * 2001-04-02 2006-03-15 Hongfen Li ANTINEOPLASTIC DRUG
JP3943399B2 (en) * 2002-01-16 2007-07-11 マイタケ プロダクツ インコーポレーテッド Fat-soluble extract from Yamabushidatake
EP1539204A1 (en) 2002-08-30 2005-06-15 Biopharmacopae Design International Inc. Plant extracts for treatment of angiogenesis and metastasis

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4613591A (en) * 1982-08-23 1986-09-23 Tsumura Juntendo Inc. Adminiculum for antitumor agents
US20030165580A1 (en) * 2002-03-04 2003-09-04 Xinxian Zhao Safe pharmaceutical composition for treatment and prevention of gynecological disease

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050191242A1 (en) * 2003-11-25 2005-09-01 Janice Brissette Foxn1 and pigmentation
US7687265B2 (en) 2003-11-25 2010-03-30 The General Hospital Corporation Foxn1 and pigmentation
US20100247627A1 (en) * 2003-11-25 2010-09-30 The General Hospital Corporation, A Massachusetts Corporation Foxn1 and pigmentation
US20110076303A1 (en) * 2009-06-25 2011-03-31 Tokuro Iwabuchi Methods for Screening for Anti-Graying Agents on the Basis of AFF-4
US9585864B2 (en) * 2010-01-05 2017-03-07 National Dong Hwa University Anticancer formulation
US20150216836A1 (en) * 2010-01-05 2015-08-06 National Dong Hwa University Anticancer formulation
US20110165201A1 (en) * 2010-01-05 2011-07-07 National Dong Hwa University Anticancer formulation
US8927601B2 (en) * 2011-12-20 2015-01-06 National Dong Hwa University Uses of N-butylidenephthalide in treating a liver injury and improving liver function
US20140212970A1 (en) * 2013-01-25 2014-07-31 China Medical University Method and kit for culturing stem cells
US9504751B2 (en) 2014-04-24 2016-11-29 Everfront Biotech Inc. Stable pharmaceutical composition
EP3338775A4 (en) * 2015-08-19 2019-04-03 Everfront Biotech Inc. Use of butylidenephthalide
US10987339B2 (en) 2015-08-19 2021-04-27 Everfront Biotech Inc. Uses of butylidenephthalide
US10688128B2 (en) * 2016-05-23 2020-06-23 Everfront Biotech Inc. Use of Z-butylidenephthalide in activating autoimmune system
CN114989891A (en) * 2022-05-25 2022-09-02 湖南诺泽生物科技有限公司 Angelica essential oil and preparation method and application thereof

Also Published As

Publication number Publication date
EP1645280A1 (en) 2006-04-12
SG121980A1 (en) 2006-05-26
TWI298259B (en) 2008-07-01
US20090111870A1 (en) 2009-04-30
US20070134351A1 (en) 2007-06-14
JP2006117663A (en) 2006-05-11
ATE455550T1 (en) 2010-02-15
TW200616656A (en) 2006-06-01
US7455861B2 (en) 2008-11-25
DE602005019010D1 (en) 2010-03-11
EP1645280B1 (en) 2010-01-20
JP4781771B2 (en) 2011-09-28

Similar Documents

Publication Publication Date Title
US7455861B2 (en) Angelicae sinensis extracts useful for treatment of cancers
Shafi et al. Induction of apoptosis in HeLa cells by chloroform fraction of seed extracts of Nigella sativa
CN1943606B (en) Chinese angelica root extract for treating cancer
TWI300352B (en) Water soluble extract from plant of solanum genus and the preparation process thereof, and pharmaceutical composition containing the water soluble extract
US10660928B2 (en) Pharmaceutical composition containing combination extracts of Moutan Root Bark, Angelica Dahurica Root, bupleurum root or fractions thereof for prevention and treatment of neurodegenerative disorder
Zhang et al. Dehydrocostus lactone inhibits cell proliferation and induces apoptosis by PI3K/Akt/Bad and ERS signalling pathway in human laryngeal carcinoma
Grech-Baran et al. Approaches of Rhodiola kirilowii and Rhodiola rosea field cultivation in Poland and their potential health benefits
CN101805246B (en) Urushiol compound and medicinal composition thereof, preparation method and application thereof
Borovskaya et al. Evaluation of the Effect of p-Tyrosol on the Level of DNA Damage in the DNA Comet Assay In Vivo
KR100458131B1 (en) Extract of Acanthopanacis Cortex having neuroprotective effects and pharmaceutical composition containing the same
US20080050426A1 (en) Method For Preparing Extract From Wild Ginseng Showing Anticancer Activity And The Composition Comprising The Same
KR101021975B1 (en) A composition for enhancing the radiotherapy of cancer
KR20200104016A (en) A pharmaceutical composition comprising compounds isolated from Phlomoides umbrosa(Turcz.) Kamelin and Makhm for preventing or treating cancer
US20110124741A1 (en) Radiosensitizer compositions comprising schisandra chinensis(turcz.)baill and methods for use
Ismeel Cytogenetic and cytotoxic studies on the effect of phytoinvestigated active compounds of Hyoscyamus niger (in vivo and ex vivo)
CN101138587A (en) Application of white fleece-flower root or white fleece-flower root diketone A in the preparation of medicament for treating neurological disorders
KR101595987B1 (en) A composition comprising Osmanthus matsumuranus extracts having anti-cancer activity
KR101675642B1 (en) A composition comprising Carpinus pubescens extracts having anti-cancer activity
Fahmi et al. Efficacy of Prunus Armeniaca on Oral Squamous cell carcinoma Cell Line: An In-Vitro Analysis of a Cell Line
Suloev et al. Isolation of individual flavonoids from Solidago canadensis L
KR101115504B1 (en) An anti-cancer composition for the prevention and treatment of carcinogenesis containing the extracts of Asparagus cochinchinensis as an active ingredient
KR101591389B1 (en) A composition comprising Litsea populifolia extracts having anti-cancer activity
KR101561736B1 (en) A composition comprising Treculia africana extracts having anti-cancer activity
US20180221329A1 (en) Cis- and trans-suffruticosol d as therapeutic agents
JP2008081412A (en) Apoptosis inducing agent

Legal Events

Date Code Title Description
AS Assignment

Owner name: BUDDHIST TZU CHI GENERAL HOSPITAL, TAIWAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NULIV SCIENCE USA, INC.;REEL/FRAME:017075/0969

Effective date: 20051206

Owner name: BUDDHIST TZU CHI GENERAL HOSPITAL, TAIWAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUO, JIANN-KUAN;HARN, HORNG-JHY;CHANG, WEN-LIANG;AND OTHERS;REEL/FRAME:017076/0021;SIGNING DATES FROM 20051110 TO 20051213

STCB Information on status: application discontinuation

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