WO2009068872A1 - Antiviral - Google Patents

Antiviral Download PDF

Info

Publication number
WO2009068872A1
WO2009068872A1 PCT/GB2008/003947 GB2008003947W WO2009068872A1 WO 2009068872 A1 WO2009068872 A1 WO 2009068872A1 GB 2008003947 W GB2008003947 W GB 2008003947W WO 2009068872 A1 WO2009068872 A1 WO 2009068872A1
Authority
WO
WIPO (PCT)
Prior art keywords
extract
astragalus
genus
virus
active fraction
Prior art date
Application number
PCT/GB2008/003947
Other languages
French (fr)
Inventor
Shouming Zhong
Hongwen Yu
Edward Blair
Original Assignee
Phynova Limited
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 Phynova Limited filed Critical Phynova Limited
Publication of WO2009068872A1 publication Critical patent/WO2009068872A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/48Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
    • A61K36/481Astragalus (milkvetch)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to an antiviral, and its use in the treatment of infections/ diseases caused by one or more genus of the flaviviridae family of viruses, including viruses from the genus: Flavivirus, Pestivirus, Unassigned Flaviviradae and Tentative Species in the Genus Hepacivirus. Inparticular, it relates to the use of an extract of Astragalus in the manufacture of a medicament for use in the treatment of Dengue Virus, West Nile Virus, Japanese Encephalitus and Yellow fever (all mosquito bourne viruses) as well as other related viruses.
  • KUN Kunjin virus
  • MVEV Murray Valley encephalitis'virus
  • SLEV St. Louis encephalitis virus
  • Flavivirus including: o Kyasanur Forest disease virus (KFD) o Langat virus (LGT) o Louping ill virus (LI) o Omsk hemorrhagic fever virus (OHF) o Powassan virus (POW) o Tick-borne encephalitis virus (TBE)
  • Pestivirus including: o Bovine viral diarrhea virus 2 (BVDV), and o Classical swine fever virus (CSFV)
  • Unassigned viruses including o GBV-A, and o GBV-C and
  • the product is composed of four herbal extracts and is formulated as follows:
  • the systematic solvent fractionation utilizes a number of different solvents commencing with the least polar and finishing with the most polar.
  • the active fraction is a dichloromethane fraction or a solvent with similar polarity.
  • the active fraction (and sub- fractions) can be characterized with reference to one or more markers, one or a combination of which may be responsible for the anti-HCV activity and polymerase inhibition.
  • markers include:
  • the plant which exhibits the activity is a member of the Leguminosae family, more particularly Huang Qi:
  • Bge var. Mongholicus Hsiao (hereafter Astragalus).
  • the plant may be referred to as Milkvetch in Europe and it is the root which is used.
  • CNl 616097 which discloses a lozenge containing attenuated Newcastle disease virus vaccine or deactivated Newcastle disease virus vaccine, 10-120 hemagglutination units, astragalus root l-20000mg, liquiritigenin 1-lOOOmg, supplementary material and stabilizer. It is stated to be suitable for preventing SARS and other viral infectious respiratory tract diseases and to have auxiliary treatment effects on hepatitis B and C.
  • CNl 607003 which discloses the use of a composition comprising Chinese caterpillar fungus (50-90 parts) with astragalus root (10-50 parts) as an auxiliary therapeutic agent for hepatitis C. It is stated to adjust immune function and improve curative effect.
  • CN 1448163 which discloses a Chinese recipe comprising astragalus root among some eleven herbs to treat various diseases including hepatitis C.
  • CN1393255 which discloses a nineteen herb mix, including astragalus root, for treating hepatitis C.
  • CN1593586 which discloses a pill, for treating hepatitis C, comprising at least 15 herbs one of which is raw astragalus root.
  • RNA • Positive-stranded, single stranded RNA including: o The genus - Flavivirus, (Table 1.1); o The genus - Pestivirus, (Table 1.2); o The genus - Unassigned Flaviviradae (Table 1.3) or o Tentative Species in the Genus HCV (Table 1.4) which may be considered separate to the HCV viruses.
  • WO 2004/112720 teaches the use if GRII antagonists (glucocorticoids), including Astragaloside II, as compounds that inhibit viral replication.
  • GRII antagonists glucocorticoids
  • Astragaloside II Astragaloside II
  • US6555523 discloses flavin compounds, including formononetin compounds, to treat viral infections.
  • Biol. Pharm. Bull vol 19(9) 1166-1169 discloses the oral administration of a decoction of astragali radix against Japenese Encephalitis virus infection in mice.
  • CN1985877 discloses the use of an astragalus extract to treat viral Hepatitis.
  • BMC Bioinformatics vol 7 (sup 5) 2006 provides a phylogenetic tree for the Flaviviridae.
  • the Astragalus extract of the invention might be used alone as an antiviral to treat one or more of the viruses listed in the tables below:
  • an Astragalus extract or an active fraction thereof or an active compound isolated therefrom for use in the manufacture of a medicament for the treatment of a disease caused by a genus of the flaviridae family selected from: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus — Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
  • the Astragalus extract or an active fraction thereof or a compound isolated therefrom are used against the following viruses: • Flavivirus o Tick-borne viruses
  • Pestivirus o Bovine viral diarrhea virus o Classical swine fever virus
  • the extract has proved capable of significantly reducing viral load in viruses from two of the four genus of flaviviridae.
  • the active fraction is an extract of Astragalus purified by at least a factor of 10 with reference to dried raw plant material, and which is characterized in that it comprises at least one marker selected from the group consisting of:
  • the extract is purified by at least a factor of 50, most preferably by a factor of between 75 and 200 and can be characterized by the presence of the four markers identified above. In fact, at least six defined peaks can be identified in this fraction.
  • the extract is used or formulated as a botanical drug.
  • the formulation is from a single herb extract.
  • the extract may be used as a food, dietary supplement or food additive.
  • the term food includes food or drink and articles used for components of such articles.
  • the raw material is cultivated and harvested from a designated production site under managed agricultural practice.
  • a third aspect of the present invention there is provided the use of Astragalus or an extract thereof or an active fraction thereof or an isolated compound therefrom in the manufacture of a food, dietary supplement or food additive for use as an antiviral to treat infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
  • the preferred method for producing an extract of Astragalus which exhibits antiviral activity and which is purified by at least a factor of 10 with reference to dried raw plant material) which is characterized in that it comprises at least one marker selected from the group consisting of:
  • the alcoholic extraction is an ethanolic extraction.
  • the ethanolic extraction is preferably conducted under reflux. Most preferably it is repeated.
  • Preferably high concentration ethanol is used (greater than 50% by concentration.)
  • the alcoholic extraction is followed by an ethanol-water precipitation and the supernatant recovered. The ethanol is recovered leaving a concentrate.
  • the concentrate is then dissolved in water to give an aqueous solution which is subjected to systematic solvent fractionation using solvents of different polarity.
  • Preferred solvents are: Petroleum ether, dichloromethane, and ethyl acetate.
  • a macro-porous resin column purification process may be employed in which the purification comprises running the extract through the column, washing with a low concentration ethanol (between 30 and 50% ethanol concentration) and eluting using a high concentration ethanol (typically 90% ethanol).
  • a method for producing an extract of Astragalus which exhibits antiviral activity against a virus from one of: o the genus - Flavivirus (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
  • the active fraction from each of the methods is referred to as an AS-C fraction due to its activity and the presence of common marker compounds.
  • the benefit of the macro- porous resin separation is its scalability and the fact it avoids the use of undesirable solvents e.g. petrol ether and dichloromethane.
  • a method of treating infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4). comprising administering an effective amount of an. Astragalus extract or an active fraction thereof or an active compound isolated therefrom to a patient or animal.
  • the Astragalus extract or an active fraction thereof or an active compound isolated therefrom is preferably provided in unit dosage form. Most preferably it is provided in a form suitable for oral delivery e.g. a filled capsule. The skilled person will appreciate other dosage forms may alternatively be presented.
  • the extract is used in an amount equivalent to an amount of between 9 and 3Og of dried raw material / day.
  • the Astragalus or an active fraction thereof is administered by way of a combination treatment with at least one other immuno-modulatory or antiviral drug e.g. Interferon and/ or Ribavirin.
  • at least one other immuno-modulatory or antiviral drug e.g. Interferon and/ or Ribavirin.
  • the drugs may be administered together simultaneously or sequentially.
  • the botanical drug, food, dietary supplement or food additive preferably comprises an extract of Astragalus purified by at least a factor of 10 with reference to dried raw plant material, and is characterized in that it comprises at least one marker selected from the group consisting of:
  • an Astragalus extract fraction having an HPLC fingerprint showing a primary peak for Astragaloside I substantially as shown in any of Figs 16a to 16d.
  • an Astragalus extract having a TLC fingerprint comprising six identifiable spots substantially as illustrated in any of lanes 2 to 5 of Fig 14 or 15.
  • Fig 1 illustrates the anti-hepatitis C virus activity of both Salvia (SMR) and Astragalus (AMR) extracts together with toxicity data;
  • Fig 2 is a flow diagram of an alcoholic extraction process giving rise to a primary extract containing less than 15% by weight of the starting raw material
  • Fig 3a is a flow diagram showing the fractionation of the primary extract (8) to give a more concentrated active fraction or secondary extract (AS-C (14));
  • Fig 3b is a flow diagram showing the fractionation of the primary extract to give a more concentrated alternative active fraction or secondary extract (ASC-90(30));
  • Fig 3 c is a flow diagram showing the fractionation of the primary extract to give a more concentrated alternative active fraction or secondary extract (ASC-90(50));
  • Fig 4 is a TLC fingerprint of a number of different fractions (UV254nm);
  • Fig 5 is a TLC fingerprint of a number of different fractions (UV365nm);
  • Fig 6 is a TLC fingerprint of a number of different fractions (daylight);
  • Fig 7 is a graph showing the activity of various fractions in a replicon assay;
  • Fig 8 is a graph showing the activity of ASC 705 (14) at different concentrations;
  • Fig 9 is a flow diagram showing the further fractionation of the secondary extract;
  • Fig 10 is a TLC fingerprint of the five fractions AS-C-I to AS-C -V in chloroform/ methanol 9:1
  • Fig 11 is a TLC fingerprint of the five fractions AS-C-I to AS-C -V in chloroform/ methanol/ water 8:2:02
  • Fig 12 is a flow diagram showing the isolation, purification and chemical identification of the components present in different AS-C-I sub fractions
  • Fig 13a is a TLC chromatogram of "ASC-I-I" in chloroform: ethanol: water 85:15:1;
  • Fig 13b is a TLC chromatogram of "ASC-I-2" and “ASC-I-3" in chloroform: ethanol: and water 85:15:1 (UV 254nm);
  • Fig 13c is a TLC chromatogram of "ASC-I-2" and "ASC-I-3" in chloroform: ethanol: water 85:15:1 (iodine);
  • Fig 14 is a TLC chromatogram under UV 254nm of ASC 90(30) and ASC 90(50) extracts;
  • Fig 15 is a TLC chromatogram under sunlight of ASC 90(30) and ASC 90(50) extracts;
  • Figs 16 a - b are HPLC fingerprints of ASC 90(30)
  • Figs 16 c - d are HPLC fingerprints of ASC 90(50) 8 003947
  • Fig 17 is a representation of the phylogenetic relationship of the flaviviridae family of viruses based on NS3 helicase region;
  • Fig 18 is representation of the phylogenetic relationship of the flaviviridae family of viruses based on E protein amino acid identity
  • Fig 19 is a diagrammatic representation of the clinical manifestations of Dengue virus infection
  • Fig 20 is a diagrammatic representation shown in factors influencing disease severity
  • Fig 21 shows graphically the cytotoxicity data generated in Example 8
  • Fig 22 show graphically the activity data generated in Example 8
  • Fig 23 shows graphically Den 2 assay data generated in Example 8.
  • Fig 1 illustrates the activity of the PYN 17 constituents:
  • Both AMR and SMR appear to demonstrate inhibitory activity in the HCV replicon assay. This is particularly evident at high concentrations, as shown in the left hand panel of Fig 1.
  • the right hand panel of Fig 1 shows cytotoxicity data in the same replicon cells. This indicates that AMR displays no observable cell cytotoxicity whereas SMR is cytotoxic at high concentrations.
  • Root material was dried in an oven at 6O 0 C for 3 hours, pulverised into a coarse powder and passed through a sieve (10 mesh). It was then subjected to an alcoholic extraction as set out in Fig 2.
  • This powdered primary extract had a solid yield of between 11.7 and 13% by weight compared to the dry weight of starting material, i.e. It had been purified by a factor of about 8.
  • the content of Astragaloside IV 5 the standard chemical marker for Astragalus was greater than 0.4 %.
  • the primary extract obtained by the method illustrated in Fig 2 was fractionated as set out in Fig 3 a.
  • Figs 4-6 are TLC plates showing from left to right, fractions (9), (10), (13), (14), (15), and (16).
  • the TLC plates are silica gels and the developing system was chloroform/ methanol/ water (8:2:0.2).
  • Fig 4 shows detection with UV at 254nm
  • Fig 5 shows detection with UV at 365nm
  • Fig 6 is observed in daylight following treatment with 10% sulphuric acid in ethanol followed by heating.
  • Fraction (14) shows a plurality of distinct spots. At least 6 are clearly seen at 254nm
  • Fraction 14 AS-C was found to be particularly active in the replicon assay (Example 4 Table 8). It had a solid yield of about 1.1% by weight compared to the dry weight of starting material. In fact it had been purified by a factor of about 90%. This fraction had a chromatographic profile as illustrated in Figs 4-6.
  • AS-C 90 (30) and AS-C 90 (50 are as follows
  • the raw material of Astragalus was pulverized and extracted twice with 70% ethanol (10 times of the weight of the raw material for 1.5 hours and then 8 times for 1 hour). The extracted solutions were combined and the ethanol recovered under vacuum to about half of the weight of the raw material. 95% ethanol was added to the concentrated extract to give an ethanol concentration of 80%. The concentrated extract was set aside over night and filtered to obtain the solution. The solvent was recovered under vacuum from the solution to dryness with a temperature not exceeding 70°C to produce a dried extract (AS-C).
  • test sample Under the sunlight, the test sample showed the same spots as the reference substances AS-C-I-I and Astragaloside IV. (Fig 15)
  • Apparatus Waters HPLC Systems: Waters 2420 ELS Detector, 717 plus Autosampler & Pump 600 Controller with EmpowerTM Software.
  • Nebulizer heater level 50% Drift tube heater set-temp: 60°C
  • Table 4 A quantitative analysis is given in Table 4 below: Table 4.
  • standardised extract is standardised to one or more of the "mean” figures, i.e. to within plus or minus 40%, more preferably plus or minus 30%, and most preferably to within plus or minus 20%.
  • the DMSO solutions were then diluted 1/10 in tissue culture medium and filtered. This concentration was called NEAT. lO ⁇ l per well was used, total volume in the test well lOO ⁇ l. All concentrations were set up as 5 replicates. 3 replicates were of the same dilution as the DMSO.
  • the replicon cells were plated out at a concentration of 5 x 10 3 / well in 90 ⁇ l. The following day the test samples were added in lO ⁇ l. The plates were incubated for a further 72 hours and harvested and assayed using the Dual Luciferase Assay Promega. The replicon cell line expresses Renilla luciferase.
  • the samples were tested for cytotoxicity by setting up a separate 96 well plate with each sample and concentration in duplicate.
  • the cells were labeled with tritiated thymidine for 24 hours and harvested.
  • Assay 1 Mean of reading from cells without any drug added 198906. The results are shown in Table 8.
  • IFN alpha concentrations are pg/ml.
  • results are illustrated graphically in Figs 7 and 8.
  • Fig 7 shows the raw replicon scores of the samples against concentration.
  • ASC-0705 is clearly the best performer and outperforms IFN alpha (standard treatment). It also demonstrates that the best activity is at a concentration above 10ug/ml.
  • the fraction (14 AS-C) was subjected to a chromatographic separation step as illustrated in Fig 9 and as set out below. Briefly this comprised the following steps:
  • Fraction 15 (18) had a weight of 241 mg and was further purified twice with Sephadex column chromatography and eluted with methanol to get a fraction (23)
  • Fraction 17 (19) had a weight of 231 mg and was further separated into two fractions by way of silica gel column chromatography eluting with chloroform/methanol (94:6). The two fractions weighed 13mg, fraction (24) "AS- C-III” and a 14mg (25) "AS-C-IV";
  • Fig 10 shows silica gel plates developed in chloroform / methanol (9:1). The left hand plates is viewed under UV at 254nm and the right hand plate under daylight (spray with 10% sulphuric acid in ethanol and then heat the plate until the spots are clearly seen).
  • Fig 11 shows silica gel plates developed in chloroform / methanol/ water (8:2:0.2). The left hand plates is viewed under UV at 254nm and the right hand plate under daylight (spray with 10% sulphuric acid in ethanol and then heat the plate until the spots are clearly seen).
  • AS-C fraction contained several (>6) small peaks (m/z 269, 301, 303, etc.)
  • AS-C-II and AS-C-IV contained a single peak which is higher than the DMSO peaks.
  • AS-C-III contains a single peak which is lower than the DMSO peak.
  • the samples were tested for anti-HCV activity in: a) a replicon assay (Reblikon), b) a NS5B assay and c) a NS3/NS4A full length protease assay.
  • Reblikon a replicon assay
  • NS5B a NS5B assay
  • NS3/NS4A full length protease assay
  • AS-C-I and AS-C-II • a) also show some activity in the replicon assay but at higher concentrations.
  • the determined EC 50 values were 36 and 17 ⁇ g/ml respectively
  • AS-C-I The precipitated white powder from the 18 th fraction (20) was referred as "AS-C-I" (649mg).
  • the ASC-I (20) was dissolved in methanol with supersonication, and then filtrated. The filtrate was separated by Sephadex LH-20 column to obtain three major compounds as showed in the TLC plates illustrated in Figs 13-15.
  • the three compounds identified were analyzed by mass spectrum and nuclear magnetic resonance spectroscopy, respectively.
  • the structures of the three compounds were identified by comparing the data obtained with the existing literature on chemical structures and are shown in Table 13.
  • Formula 2 Formononetin-7-O- ⁇ -D-glucoside (AS-C-I-2)
  • the objective was to test: a) The cytotoxicity of extracts ASC, ASC 90(30) and ASC 90(5O) 5 and b) Test the effects of extracts ASC, ASC 90(30) and ASC 90(50) on dengue virus infection METHOD
  • test sample (ASC 0506, ASC 90 (30) and ASC 90 (50) (10 mg) was re-suspended in 1 ml of DMSO to obtain the concentration of 10 mg/ml.
  • the completely dissolved extract was diluted by a 100-fold with growth medium and, therefore, this stock solution had the final concentration of 100 ⁇ g/ml (in the presence of 1% DMSO).
  • the stock solution was serially diluted with the maintenance medium as follows.
  • Vero an African green monkey kidney cell line
  • Eahy 926 an endothelial cell line
  • Vero an African green monkey kidney cell line
  • Eahy 926 an endothelial cell line
  • Vero cells were seeded onto 96-well plates (20,000 cells/ well) in 100 ⁇ l of growth medium (10% FBS-MEM + 2 niM L-glutamine for Vero cells and 10% FBS-DMEM F-12 for Eahy 926) and incubated at 37 0 C with 5% CO 2 for 24 hr. Thereafter, the medium was discarded and the cells were incubated with 100 ⁇ l of extracts ASC, ASC 90(30), or ASC 90(50) at the concentrations of 0.2, 0.5, 2, 5, 20, 50, and 100 ⁇ g/ ⁇ l.
  • Standard controls for cell proliferation at the indicated time points were also established by plating different numbers of cells in each well: 10 4 , 105, 10 6 cells/well for Vero or 10 3 , 10 4 , 5 x 10 4 , 10 5 cells/well for Eahy 926.
  • the absotbance which reflects the number of cells was read out at 595 run (with the reference wavelength of 630 nm).
  • the numbers of DMSO or compound-treated cells were obtained from the predicted value on the standard curve [OD (standard - blank) vs. the number of cells].
  • the percentage of cell viability was calculated based on the following formula:
  • % viability The number of DMSO or compound-treated cells x 100
  • Vero or Eahy 926 cells were seeded onto 24-well plates (250,000 cells/well) in 1 ml of growth medium (10% FBS-MEM + 2 mM L-glutamine) and incubated at 37 0 C with 5% CO 2 for 24 hr. Thereafter, the medium was discarded and cells were incubated with dengue virus serotype 2 (strain 16681) at an MOI of 1 or 5 in the total volume of 250 ⁇ l at 37°C for 2 hr.
  • growth medium (10% FBS-MEM + 2 mM L-glutamine
  • the virus supernatant was then removed and the cells were washed twice with 1 ml of plain medium and further incubated with 1.25 ml of maintenance medium (2% FBS-MEM + 2 mM L-glutamine) which contains compounds ASC, ASC 90(30) or ASC 90(50) at the concentration of 5 or 50 ⁇ g/ ⁇ l, or DMSO (the diluent of the compounds) at the concentration of 0.05% or 0.5%.
  • maintenance medium 2% FBS-MEM + 2 mM L-glutamine
  • DMSO the diluent of the compounds
  • the percentage of dengue virus infection was assessed by immunofluorescence staining of intracellular dengue viral E and NSl and subsequent analysis by flow cytometry.
  • titers of infectious virus in the culture supernatants were determined at the indicated time points by a focus forming unit assay.
  • Vero and Eahy 926 cells were cultured in the presence of extracts ASC, ASC 90(30), and ASC 90(50) at the concentrations of 0.2, 0.5, 2, 5, 20, 50, and 100 ⁇ g/ml up to 3 days.
  • the extract-treated cells were harvested daily and determined for the viability by an MTT assay based on cell proliferation.
  • DMSO-treated cells collected at the same time points served as the control. Results from two independent experiments showed that all extracts did not cause significantly cell death in both cell lines tested but, instead, were likely to induce cell proliferation as compared with the control. In addition, cell viability appeared to be time-dependent since it became decreased on day 3 after culture establishment.
  • Vero and Eahy 926 cells were incubated with dengue virus serotype 2 (strain 16681) at an MOI of 1 or 5 for 2 hr and thereafter the virus supernatant was replaced with fresh medium containing extracts ASC, ASC 90(30), or ASC 90(50) at the final concentrations of 5 or 50 ⁇ g/ml. The cultures were further incubated up to 3 days. Mock and dengue virus-infected cells that had been treated or left untreated with DMSO served as controls for dengue virus infection.
  • treatment with extracts at the concentration of 5 ⁇ g/ml yielded the similar percentage of viable cells as compared with DMSO-treated control.
  • the viability of dengue virus-infected cells tended to decrease in the presence of 50 ⁇ g/ml of the extracts, particularly ASC and ACS 90(50), as determined by tryptan blue and propidium iodide staining.
  • Dengue virus infection resulted in the expression of viral E and NSl antigens in Vero cells in a dose-dependent and a time-dependent manner (Fig 22) and almost all of the cells (about 90%) could be infected with dengue virus by day 3 post infection.
  • treatment with the extracts at 5 ⁇ g/ml did not affect the percentage of viral antigen expression as well as the production of infectious virus as assessed by immunofluorescence staining and a focus forming unit assay, respectively.
  • Extracts ASC, ASC 90(50) and ASC 90(30) at all the concentrated tested were not toxic to Vero and Eahy 926 cells in the absence of dengue virus infection.
  • the predicted CC50 of each extract may therefore be higher than 100 ⁇ g/ml.
  • treatment with 50 ⁇ g/ml of the ASC and ASC 90(50) resulted in certain extents of cell death in dengue virus-infected Vero and Eahy 926 cultures. This phenomenon was coincident with a slightly reduction of viral antigen expression and a significant decrease in virus production in the same cultures following treatment with these extracts.
  • the induction of cell death by the extracts ASC and ASC 90(50) may be one of the potential mechanisms to reduce the initial pool of infectious viruses during dengue virus infection of the susceptible cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Botany (AREA)
  • Biotechnology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Medical Informatics (AREA)
  • Engineering & Computer Science (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Oncology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

The present invention relates to a novel antiviral product and its use in the treatment of the flaviviridae family of viruses including the genus flavivirus, particularly Dengue.

Description

ANTIVIRAL TECHNICAL FIELD OF THE INVENTION
The present invention relates to an antiviral, and its use in the treatment of infections/ diseases caused by one or more genus of the flaviviridae family of viruses, including viruses from the genus: Flavivirus, Pestivirus, Unassigned Flaviviradae and Tentative Species in the Genus Hepacivirus. Inparticular, it relates to the use of an extract of Astragalus in the manufacture of a medicament for use in the treatment of Dengue Virus, West Nile Virus, Japanese Encephalitus and Yellow fever (all mosquito bourne viruses) as well as other related viruses.
The applicant has previously described in UK application number 0612025.7, which at the time of filing remains unpublished, that an Astragalus extract, and defined fractions therof, exhibit antiviral activity against Hepatitis C virus (HCV) in:
• A replicon test,
• A polymerase test (NS5B), and
• Protease tests (NS3/4A).
Based on, particularly, the polymerase test data they suggested that the Astragalus extract and defined fractions thereof might exhibit activity against other viruses within the Flaviviradae family of viruses.
Subsequently, they have demonstrated, as set out herein, that the Astragalus extract disclosed in the earlier application, together with a variant thereof, are active in reducing viral load against Dengue viruses and that as a consequence of the phylogenetic relationship of the Flaviviruses based on the NS3 helicase region (Fig 17) and E protein amino acid identity (Fig 18), together with the activity previously demonstrated against HCV, argue that this provides credible evidence that extracts of Astragalus, and defined fractions thereof, including single compounds isolated therefrom, are likely to exhibit activity against the Flaviviridae identified in Tables 1.1 to 1.4 herein. In particular the evidence supports the use of extracts of Astragalus in the manufacture of a medicament for use in the treatment of at least the following mosquito bourne Flaviviruses:
• Dengue Virus (DEN);
• West Nile (WN) and Japanese Encephalitis (JE), two very closely related viruses;
• Kunjin virus (KUN), Murray Valley encephalitis'virus (MVEV), St. Louis encephalitis virus (SLEV); and
• Yellow fever.
Additionally, there is strong phylogenetic evidence suggesting the extract will also be active against:
• Tick bourne Flavivirus including: o Kyasanur Forest disease virus (KFD) o Langat virus (LGT) o Louping ill virus (LI) o Omsk hemorrhagic fever virus (OHF) o Powassan virus (POW) o Tick-borne encephalitis virus (TBE)
• Pestivirus including: o Bovine viral diarrhea virus 2 (BVDV), and o Classical swine fever virus (CSFV)
• Unassigned viruses including o GBV-A, and o GBV-C and
• Tentative Species in the Genus Hepacivirus, including o GBV-B BACKGROUND OF THE EWENTION
In applicant's international application WO2005079823 there is disclosed a four herb combination product which in addition to providing symptomatic relief had been shown, by way of a replicon assay, to exhibit anti-HCV activity (inhibition of 41.8 %) at a dilution of 1/350.
The product is composed of four herbal extracts and is formulated as follows:
Herbal extracts:
• Milk Thistle Fruit dry extract 0.200 g
• Chinese Sage Root dry extract 0.225 g
• Schisandra Fruit dry extract 0.400 g
• Astralagus Root dry extract 0.585 g
Excipients:
• Macrogol 6000 powder 0.600 g
• Ferwogel 30.385 (molecular weight
3.5-4.O xIO6) 0.07Og
• Mannitol EZ 0.16Og
• Aerosil 200 0.050g
• Aspartame 0.050 g
• Caramel powder 0.100 g
• Peppermint powder aroma 0.06Og
The applicant investigated this product further and surprisingly discovered that an anti- HCV activity appeared to derive from only one plant, demonstrating both good activity and no detectable cell cytotoxicity. This discovery forms the basis of UK patent application no 0612025.7, as yet unpublished. That plant is Astragalus, and it has further been determined that a fraction purified by a factor of greater than 10, more particularly greater than 50, more particularly still greater than 75 and most particularly between 75 and 200 is particularly active.
The active fraction can be produced in good yield in effectively a three stage process comprising:
1) An alcoholic extraction (which may be repeated);
2) An ethanol- water precipitation process (which may be repeated); and
3) A systematic solvent fractionation step with a plurality of solvents of different polarity.
Preferably, the systematic solvent fractionation utilizes a number of different solvents commencing with the least polar and finishing with the most polar. The active fraction is a dichloromethane fraction or a solvent with similar polarity.
The active fraction (and sub- fractions) can be characterized with reference to one or more markers, one or a combination of which may be responsible for the anti-HCV activity and polymerase inhibition. Particular markers identified include:
• Astragaloside I,
• Astragaloside IV,
• Formononetin-7-O-β-D-glucoside; and
• 3'-hydroxyl- formononetin -7-O-β-D-glucoside.
Additionally they can be identified by way of TLC or HPLC fingerprints as set out in the detailed description.
The plant which exhibits the activity is a member of the Leguminosae family, more particularly Huang Qi:
Pharmaceutical name: Radix Astragali Membranaceous;
Botanical name: Astragalus membranaceus (Fisch) Bge. or Astragalus membranaceus
Bge var. Mongholicus Hsiao (hereafter Astragalus). The plant may be referred to as Milkvetch in Europe and it is the root which is used.
In traditional Chinese herbal medicine a dosage (based on dry raw material) of 9-3Og and occasionally up to 6Og is used. Typically it is taken as a decoction. According to Pharmacopoeia of the People's Republic of China (English Edition 2000) VoI I a cold water extraction method gives a water soluable extract of not less than 17%.
According to Chinese Herbal Medicine, Materia Medica, Revised edition, chemically the root of Astragalus membranaceus is known to contain, as major ingredients, D-β asparagine, 2'4'-dihydroxy-5, 6-dimethoxyisoflavane, calycosin, formononetin, cycloastragenol, astragalosides, choline, betaine, kumatakenin, sucrose, glucuronic acid and β-sitosterol.
Its chemical constituents are discussed further in Chinese Drugs of Plant Origin, Springer- Verlag which further discusses it's pharmacology and the symptomatic relief in the treatment of chronic hepatitis.
According to Chang HM and But PP (1987) Pharmacology and applications of Chinese Materia Medica (VoI ii) World Scientific Publishing, Astragalus membranaceus may enhance immunological function. More particularly reference is made to the use of a 100% decoction, 0.4ml daily, to "protect" the liver and a clinical trial in which Huangqi injection achieved normalization of GPT levels in 80% of patients. (This is a symptomatic treatment.)
Additional art disclosing the use of Astragalus include:
CNl 616097 which discloses a lozenge containing attenuated Newcastle disease virus vaccine or deactivated Newcastle disease virus vaccine, 10-120 hemagglutination units, astragalus root l-20000mg, liquiritigenin 1-lOOOmg, supplementary material and stabilizer. It is stated to be suitable for preventing SARS and other viral infectious respiratory tract diseases and to have auxiliary treatment effects on hepatitis B and C. CNl 607003 which discloses the use of a composition comprising Chinese caterpillar fungus (50-90 parts) with astragalus root (10-50 parts) as an auxiliary therapeutic agent for hepatitis C. It is stated to adjust immune function and improve curative effect.
US2005/0074428 which discloses an adjuvant agent for use in combination with interferon and ribivarin for treating Hepatitis C containing 50-90wt % cordyceps sinensis and 10-50wt% astragalus membrancens.
CN 1448163 which discloses a Chinese recipe comprising astragalus root among some eleven herbs to treat various diseases including hepatitis C.
CN1393255 which discloses a nineteen herb mix, including astragalus root, for treating hepatitis C.
CN1593586 which discloses a pill, for treating hepatitis C, comprising at least 15 herbs one of which is raw astragalus root.
US2005/0147699 which discloses the use of an astragalus radix and codonopsis pilosulae radix mixed extract for inhibiting carcinogenesis and metastasis. It states that Astragalus is recorded, in the traditional pharmacopea, to treat chronic nephritis, albuminuria, myositis, antihypertensive, coronary artery disease, cerebral infarction, peptic ulcer (duodenal and gastric ulcer), renal disease and diabetes mellitus. It makes no reference to its potential use as an anti viral agent.
Indeed none of the documents suggests that the herb Astragalus extract can be used alone as an antiviral to treat:
• Positive-stranded, single stranded RNA including: o The genus - Flavivirus, (Table 1.1); o The genus - Pestivirus, (Table 1.2); o The genus - Unassigned Flaviviradae (Table 1.3) or o Tentative Species in the Genus HCV (Table 1.4) which may be considered separate to the HCV viruses.
WO 2004/112720 teaches the use if GRII antagonists (glucocorticoids), including Astragaloside II, as compounds that inhibit viral replication.
US6555523 discloses flavin compounds, including formononetin compounds, to treat viral infections.
Both of the above relate to the use of single compounds rather than extracts comprising a plurality of components, several of which may exhibit activity and which together provide a beneficial effect.
On the other hand, Biol. Pharm. Bull vol 19(9) 1166-1169 discloses the oral administration of a decoction of astragali radix against Japenese Encephalitis virus infection in mice.
CN1985877 discloses the use of an astragalus extract to treat viral Hepatitis.
BMC Bioinformatics vol 7 (sup 5) 2006 provides a phylogenetic tree for the Flaviviridae.
In contrast, the Astragalus extract of the invention might be used alone as an antiviral to treat one or more of the viruses listed in the tables below:
Table 1.1 Flavivirus
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Table 1.3 Unassigned viruses
Figure imgf000015_0001
Table 1.4 hepacivirus
Figure imgf000015_0002
Figure imgf000016_0001
Whilst in UK application no 0612025.7 it is stated that the data generated against HCV suggest a broader application of the extracts to treat diseases caused by the Flaviviridae, no additional test data supporting this supposition had been generated.
The applicant has since generated additional data supporting the use of an. Astragalus extract against another single-stranded RNA-containing virus, and a member of the different genus flavivirus, (particularly Dengue) which data supports the broader specific claims made herein.
SUMMARY OF THE INVENTION
According to a first aspect of the present invention there is provided an Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament for the treatment of a disease caused by a genus of the flaviridae family selected from: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus — Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
In a most preferred embodiment the Astragalus extract or an active fraction thereof or a compound isolated therefrom are used against the following viruses: • Flavivirus o Tick-borne viruses
Kyasanur Forest disease virus;
Langat virus
Louping ill virus
Omsk hemorrhagic fever virus Powassan virus
Tick-borne encephalitis virus o Mosquito-borne viruses
Dengue virus
Japanese encephalitis virus
Murray Valley encephalitis virus
St. Louis encephalitis virus
West Nile virus
Kunjin virus
Yellow fever virus
• Pestivirus o Bovine viral diarrhea virus o Classical swine fever virus
• Unassigned viruses o GB virus A o GB virus C
• Hepacivirus o GB virus B
The extract has proved capable of significantly reducing viral load in viruses from two of the four genus of flaviviridae.
Preferably the active fraction is an extract of Astragalus purified by at least a factor of 10 with reference to dried raw plant material, and which is characterized in that it comprises at least one marker selected from the group consisting of:
• Astragaloside I;
• Astragaloside IV;
• Formononetin-7-o-β-d-glucoside; and
• 3 ' -hydroxyl-formononetin-7-o-β-d-glucoside.
Preferably it comprises all four markers. In a particularly favoured embodiment, the extract is purified by at least a factor of 50, most preferably by a factor of between 75 and 200 and can be characterized by the presence of the four markers identified above. In fact, at least six defined peaks can be identified in this fraction.
In a preferred embodiment the extract is used or formulated as a botanical drug.
Most preferably the formulation is from a single herb extract.
In an alternative embodiment the extract may be used as a food, dietary supplement or food additive. The term food includes food or drink and articles used for components of such articles.
The botanical terms used herein are intended to have the meanings as used by the FDA in their directive "Guidance for Industry - Botanical Drug Products" (June 2004) which directive is incorporated by reference. It will, however, by understood by persons skilled in the art that in different countries different terminology may be used. Reference to the FDA guidance terminology is used for consistency and should not be taken to be limiting. Thus differing but equivalent terms used by, for example, the EMEA in their Guidelines on Quality of Herbal Medicinal Products / Traditional Herbal Medicinal Products (CHMP/ THMP adopted March 2006), will be understood by the skilled person to be encompassed by the terms used herein.
According to a second aspect of the present invention there is provided Astragalus raw material packaged or otherwise sold in a manner which indicates it is suitable and intended for use as an antiviral to treat infections caused by a virus from one of: o the genus - Flavivrus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4). Preferably the raw material is cultivated and harvested from a designated production site under managed agricultural practice.
According to a third aspect of the present invention there is provided the use of Astragalus or an extract thereof or an active fraction thereof or an isolated compound therefrom in the manufacture of a food, dietary supplement or food additive for use as an antiviral to treat infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
The preferred method for producing an extract of Astragalus (which exhibits antiviral activity and which is purified by at least a factor of 10 with reference to dried raw plant material) which is characterized in that it comprises at least one marker selected from the group consisting of:
• Astragaloside I;
• Astragaloside IV
• Formononetin-7-o-β-d-glucoside; and
• 3 '-hydroxyl-formononetin-7-o-β-d-glucoside comprises:
1) an alcoholic extraction (which may be repeated);
2) an ethanol-water precipitation process (which may be repeated); and either
3.1) a systematic solvent fractionation step with a plurality of solvents of different polarity or
3.2) a macro-porous resin column purification.
Preferably, the alcoholic extraction is an ethanolic extraction. The ethanolic extraction is preferably conducted under reflux. Most preferably it is repeated. Preferably high concentration ethanol is used (greater than 50% by concentration.) The alcoholic extraction is followed by an ethanol-water precipitation and the supernatant recovered. The ethanol is recovered leaving a concentrate.
The concentrate is then dissolved in water to give an aqueous solution which is subjected to systematic solvent fractionation using solvents of different polarity. Preferred solvents are: Petroleum ether, dichloromethane, and ethyl acetate.
Alternatively a macro-porous resin column purification process may be employed in which the purification comprises running the extract through the column, washing with a low concentration ethanol (between 30 and 50% ethanol concentration) and eluting using a high concentration ethanol (typically 90% ethanol).
According to a forth aspect of the present invention there is provided a method for producing an extract of Astragalus which exhibits antiviral activity against a virus from one of: o the genus - Flavivirus (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4). and which is purified by at least a factor of 10 with reference to dried raw plant material, and which is characterized in that it comprises at least one marker selected from the group consisting of: o Astragaloside I; o Astragaloside IV o Formononetin-7-o-β-d-glucoside; and o 3 '-hydroxyl-formononetin-7-o-β-d-glucoside comprising:
1) an alcoholic extraction (which may be repeated);
2) an ethanol-water precipitation process (which may be repeated); and either 3.1) a systematic solvent fractionation step with a plurality of solvents of different polarity, or
3.2) a macro-porous resin column purification.
The active fraction from each of the methods is referred to as an AS-C fraction due to its activity and the presence of common marker compounds. The benefit of the macro- porous resin separation is its scalability and the fact it avoids the use of undesirable solvents e.g. petrol ether and dichloromethane.
According to a fifth aspect of the present invention there is provided a method of treating infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4). comprising administering an effective amount of an. Astragalus extract or an active fraction thereof or an active compound isolated therefrom to a patient or animal.
The Astragalus extract or an active fraction thereof or an active compound isolated therefrom is preferably provided in unit dosage form. Most preferably it is provided in a form suitable for oral delivery e.g. a filled capsule. The skilled person will appreciate other dosage forms may alternatively be presented.
Preferably the extract is used in an amount equivalent to an amount of between 9 and 3Og of dried raw material / day.
In one embodiment the Astragalus or an active fraction thereof is administered by way of a combination treatment with at least one other immuno-modulatory or antiviral drug e.g. Interferon and/ or Ribavirin.
The drugs may be administered together simultaneously or sequentially. The botanical drug, food, dietary supplement or food additive preferably comprises an extract of Astragalus purified by at least a factor of 10 with reference to dried raw plant material, and is characterized in that it comprises at least one marker selected from the group consisting of:
• Astragaloside I;
• Astragaloside IV
• Formononetin-7-o-β-d-glucoside; and
• 3 ' -hydroxy l-formononetin-7-o-β-d-glucoside.
It may be further characterized by the presence of characteristic spots present by way of a TLC fingerprint as shown in any one of Figs 4-6 or by the characteristic peaks present by way of a HPLC chromatogram as shown in any of Figs 16a-16d.
According to a sixth aspect of the present invention there is provided an Astragalus extract fraction having an HPLC fingerprint showing a primary peak for Astragaloside I substantially as shown in any of Figs 16a to 16d.
According to a seventh aspect of the present invention there is provided an Astragalus extract having a TLC fingerprint comprising six identifiable spots substantially as illustrated in any of lanes 2 to 5 of Fig 14 or 15.
The present invention will be further illustrated, by way of Example only, with reference to the following methodology and data in which:
Fig 1 illustrates the anti-hepatitis C virus activity of both Salvia (SMR) and Astragalus (AMR) extracts together with toxicity data;
Fig 2 is a flow diagram of an alcoholic extraction process giving rise to a primary extract containing less than 15% by weight of the starting raw material;
Fig 3a is a flow diagram showing the fractionation of the primary extract (8) to give a more concentrated active fraction or secondary extract (AS-C (14)); Fig 3b is a flow diagram showing the fractionation of the primary extract to give a more concentrated alternative active fraction or secondary extract (ASC-90(30));
Fig 3 c is a flow diagram showing the fractionation of the primary extract to give a more concentrated alternative active fraction or secondary extract (ASC-90(50));
Fig 4 is a TLC fingerprint of a number of different fractions (UV254nm); Fig 5 is a TLC fingerprint of a number of different fractions (UV365nm); Fig 6 is a TLC fingerprint of a number of different fractions (daylight); Fig 7 is a graph showing the activity of various fractions in a replicon assay; Fig 8 is a graph showing the activity of ASC 705 (14) at different concentrations; Fig 9 is a flow diagram showing the further fractionation of the secondary extract;
Fig 10 is a TLC fingerprint of the five fractions AS-C-I to AS-C -V in chloroform/ methanol 9:1
Fig 11 is a TLC fingerprint of the five fractions AS-C-I to AS-C -V in chloroform/ methanol/ water 8:2:02
Fig 12 is a flow diagram showing the isolation, purification and chemical identification of the components present in different AS-C-I sub fractions
Fig 13a is a TLC chromatogram of "ASC-I-I" in chloroform: ethanol: water 85:15:1;
Fig 13b is a TLC chromatogram of "ASC-I-2" and "ASC-I-3" in chloroform: ethanol: and water 85:15:1 (UV 254nm);
Fig 13c is a TLC chromatogram of "ASC-I-2" and "ASC-I-3" in chloroform: ethanol: water 85:15:1 (iodine);
Fig 14 is a TLC chromatogram under UV 254nm of ASC 90(30) and ASC 90(50) extracts;
Fig 15 is a TLC chromatogram under sunlight of ASC 90(30) and ASC 90(50) extracts;
Figs 16 a - b are HPLC fingerprints of ASC 90(30) Figs 16 c - d are HPLC fingerprints of ASC 90(50) 8 003947
Fig 17 is a representation of the phylogenetic relationship of the flaviviridae family of viruses based on NS3 helicase region;
Fig 18 is representation of the phylogenetic relationship of the flaviviridae family of viruses based on E protein amino acid identity;
Fig 19 is a diagrammatic representation of the clinical manifestations of Dengue virus infection;
Fig 20 is a diagrammatic representation shown in factors influencing disease severity;
Fig 21 shows graphically the cytotoxicity data generated in Example 8; Fig 22 show graphically the activity data generated in Example 8; and Fig 23 shows graphically Den 2 assay data generated in Example 8.
DETAILED DESCRIPTION
Whilst the current application focuses on the non - HCV applications of the Astragalus extract, the methodology and data disclosed in UK patent no 0612025.7 are included for reference.
METHODOLOGY
EXAMPLE 1
Activity of component herbs
Following on from the discovery that the four herb combination demonstrated anti hepatitis C activity (by replicon Assay) the Applicant looked at the activity of the individual herbs. Fig 1 illustrates the activity of the PYN 17 constituents:
• AMR - Astragalus membranaceus root;
• SCF - Schisandra chinensis fruit;
• SMR - Salvia miltiorrhiza root;
• SMF - Silybum marianum fruit; and
• BCSMRO404 - A purified SMR compared to the control: • PYNl 7 - four herb combination.
Both AMR and SMR appear to demonstrate inhibitory activity in the HCV replicon assay. This is particularly evident at high concentrations, as shown in the left hand panel of Fig 1. The right hand panel of Fig 1 shows cytotoxicity data in the same replicon cells. This indicates that AMR displays no observable cell cytotoxicity whereas SMR is cytotoxic at high concentrations.
Having determined that the Astragalus membranaceus demonstrated activity and a lack of toxicity they sought to identify specific active fractions.
EXAMPLE 2.
Astragalus Root Extraction Process
Root material was dried in an oven at 6O0C for 3 hours, pulverised into a coarse powder and passed through a sieve (10 mesh). It was then subjected to an alcoholic extraction as set out in Fig 2.
Briefly this comprised the following steps:
1. To 10Og of coarse powdered Astragalus membranaceus (1 ) was added 70% ethanol (10 folds of raw material). This was then refluxed for 1.5 hours and the solution (2a) separated from the residue (3a);
2. To the residue was added 70% ethanol (8 folds of raw material). This was then refluxed for 1 hour and again the solution (2b) was separated from the residue (3b);
3. The solutions (2a and 2b) were combined, and the ethanol recovered under vacuum at a pressure of 0.08MPa to give 50ml of a concentrate (4);
4. To the concentrate was added 95% ethanol. The solution was allowed to stand over night and the precipitate separated from the supernatant (5) by filtration; 5. Again the ethanol was recovered under vacuum at a pressure of 0.08MPa and a concentrate (6) obtained;
6. The concentrate was dried at 60-70°C under vacuum at a pressure of 0.08MPa to give a solid extract (7).
7. The solid extract was then ground to a powder (8). This powdered primary extract had a solid yield of between 11.7 and 13% by weight compared to the dry weight of starting material, i.e. It had been purified by a factor of about 8.
The content of Astragaloside IV5 the standard chemical marker for Astragalus was greater than 0.4 %.
EXAMPLE 3
Extract purification/ extraction / characterisation
The primary extract obtained by the method illustrated in Fig 2 was fractionated as set out in Fig 3 a.
Briefly this comprised the following steps:
1. To the primary extract (8) was added 95% ethanol to 80% and this was again allowed to stand over night. The precipitate (9) "AS-A" was separated from the supernatant (10) "AS-F" by filtration;
2. The ethanol was recovered under vacuum at a pressure of 0.08MPa and a concentrated solution (11) obtained;
3. Water was added to the concentrated solution (11) to form a water solution (12) which was successively partitioned with: a. Petroleum ether to give a fraction (13) "AS-B"; b. Dichloromethane to give a fraction (14) "AS-C"; and c. Ethyl acetate to give a fraction (15) "AS-D"; together with d. The Water faction (16) "AS-E". The fractions (9), (10), (13), (14), (15) and (16) were subjected to further activity tests as set out in Table 2 below:
Table 2.
Sample Source Total Starch (%) Fraction codes weight(g) Yield (%)
AS-A (9) precipitates 129.0 0 12.9
AS-F (10) 80% ethanol 4.0 25 12.4 soluble fraction
AS-B (13) petroleum ether 2.0 0 0.21 fraction
AS-C (14) methylene 10.5 0 1.1 dichloride fraction
AS-D (15) ethyl acetate 1.1 0 0.12 fraction
AS-E (16) aqueous fraction 100.0 0 10.5
Figs 4-6 are TLC plates showing from left to right, fractions (9), (10), (13), (14), (15), and (16). The TLC plates are silica gels and the developing system was chloroform/ methanol/ water (8:2:0.2).
Fig 4 shows detection with UV at 254nm; and
Fig 5 shows detection with UV at 365nm;
Fig 6 is observed in daylight following treatment with 10% sulphuric acid in ethanol followed by heating.
Fraction (14) shows a plurality of distinct spots. At least 6 are clearly seen at 254nm
Fraction 14 AS-C was found to be particularly active in the replicon assay (Example 4 Table 8). It had a solid yield of about 1.1% by weight compared to the dry weight of starting material. In fact it had been purified by a factor of about 90%. This fraction had a chromatographic profile as illustrated in Figs 4-6.
In alternative protocols, Figs 3b and 3 c, the powder extract (8) from Fig 2 is treated as follows:
1. Add water (two fold) and sonicate to dissolve the powder;
2. Filter and keep the supernatant;
3. Add to a column filled with a macroporus resin;
4. Elute sequentially with either: a. 3BV water, 3BV 30% ethanol, 3BV 90% ethanol and concentrate (Fig 3b) or b. 3BV water, 3BV 50% ethanol, 3BV 90% ethanol (Fig 3c) and concentrate.
More specifically the preferred extraction processes for AS-C 90 (30) and AS-C 90 (50 are as follows
Extraction:
The raw material of Astragalus was pulverized and extracted twice with 70% ethanol (10 times of the weight of the raw material for 1.5 hours and then 8 times for 1 hour). The extracted solutions were combined and the ethanol recovered under vacuum to about half of the weight of the raw material. 95% ethanol was added to the concentrated extract to give an ethanol concentration of 80%. The concentrated extract was set aside over night and filtered to obtain the solution. The solvent was recovered under vacuum from the solution to dryness with a temperature not exceeding 70°C to produce a dried extract (AS-C).
Purification through macro porous resin Production of AS-C 90 (30):
Dissolve the dried extract in water (twice the amount of the starting raw material) , sonicate and filter. Apply the solution to a column filled with pre-treated macro porous resin. The quantity of the resin in the column was about 9 times (volume in 95% ethanol/raw material weight) of the starting raw material. Elute the column with purified water (about 3 times the column volume) until sugar test became negative. Continue the elution with 30% ethanol (about 3 times the column volume). Change the eluting solvent to 90% ethanol and continue the elution until 3 times of the column volume ethanol was used. Collect the 90% ethanol elution portion and recover the solvent under vacuum to dryness to obtain AS-C 90 (30). (Fig 3b)
Production of AS-C 90 (50):
Dissolve the dried extract in water (twice the amount of the starting raw material), sonicate and filter. Apply the solution to a column filled with pre-treated macro porous resin. The quantity of the resin in the column was about 9 times (volume in 95% ethanol/raw material weight) of the starting raw material. Elute the column with purified water (about 3 times the column volume) until sugar test became negative. Continue the elution with 50% ethanol (about 3 times the column volume). Change the eluting solvent to 90% ethanol and continue the elution until 3 times of the column volume ethanol was used. Collect the 90% ethanol elution portion and recover the solvent under vacuum to dryness to obtain AS-C 90 (50). (Fig 3c)
The resulting extracts have an appearance, by TLC, as illustrated in Figs 14 and 15 when examined using the following methodology:
TLC Method and Chromatogram
1. TLC Method
Take adequate amount of the reference substances of AS-C-I-I, AS-C-I-2 and Astragaloside IV respectively, put them into a flask and dissolve with methanol as the mixed reference solution (lmg/ml of each substance). Take adequate amount of each test sample, respectively, dissolve them with methanol - chloroform mixture (6:1) solution as the sample solution with a concentration of 2.5mg/ml, respectively. Apply lOμl of each test samples solution together with the reference solution onto a TLC plate (254nm fluorescent). Develop the plate with chloroform - methanol - water (8: 2: 0.2). Dry the plate and then spray with 10% sulphuric acid-ethanol solution. Heat with hot air blow until the spots became clear.
Under UV light (254nm), the test samples showed the same spot as the reference substance AS-C-I-2. (Fig 14)
Under the sunlight, the test sample showed the same spots as the reference substances AS-C-I-I and Astragaloside IV. (Fig 15)
In each of Figs 14 and 15 the sample order is:
1. Mixed reference substances of Astragaloside I (AS-C-I-I), Formononetin-7-O-β-D- glucoside (AS-C-I-2) and Astragaloside IV
2. Test sample batch No. 0412 , AS-C-90 ( 30 ) Fraction
3. Test sample batch No. 0604 , AS-C-90 ( 30 ) Fraction
4. Test sample batch No. 0412 , AS-C-90 ( 50 ) Fraction
5. Test sample batch No. 0604 , AS-C-90 ( 50 ) Fraction
By HPLC chromatography the extracts appear as illustrated in Figs 16a- 16d.
HPLC Method & Chromatogram
1. HPLC Detection Method
Apparatus: Waters HPLC Systems: Waters 2420 ELS Detector, 717 plus Autosampler & Pump 600 Controller with Empower™ Software.
Chromatographic Conditions:
Column: Atlantis ® dC18 ( 5μm , 250χ4.6mm )
Mobile Phase: % A acetonitrile % B water - acetic acid = 200:1 Gradient Settings: As set out in Table 3: Table 3
Figure imgf000031_0001
Flow rate: lml/min
Chromatographic column temperature : 25°C
Gain: 20 Gas pressure: 20
Nebulizer heater level: 50% Drift tube heater set-temp: 60°C
Preparation of Sample Solution
Take precisely 20mg of the sample, put it into a 10ml flask and dissolve it with 80% ethanol. Ultra-sonicate for 5 minutes and add solvent to the volume, shake it well and cool it down; filter it with 0.45μl microporous filtering film to obtain the sample solution. Load lOμl of the sample for testing.
In Figs 16 a-d the peaks can be identified as follows:
Peak Index:
1. Formononetin-7-O-β-D-glucoside (AS-C-I-2 )
2. Astragaloside IV
3. Astragaloside I (AS-C-I-I)
A quantitative analysis is given in Table 4 below: Table 4. Preferably, and standardised extract is standardised to one or more of the "mean" figures, i.e. to within plus or minus 40%, more preferably plus or minus 30%, and most preferably to within plus or minus 20%.
Most preferably standardisation is against the largest component, namely, Astragaloside
EXAMPLE 4 Activity 1
Figure imgf000032_0001
In order to test the activity of the fractions ASA to ASF lOmg of each sample was dissolved in 1 ml of DMSO and sonicated for 15 minutes. Two additional samples PA (crude Astragalus extract as WO2005079823) and INFα (standard treatment) were run as comparators. The solubility of the samples is shown in Table 5 below:
Table 5.
Figure imgf000032_0002
Figure imgf000033_0001
The DMSO solutions were then diluted 1/10 in tissue culture medium and filtered. This concentration was called NEAT. lOμl per well was used, total volume in the test well lOOμl. All concentrations were set up as 5 replicates. 3 replicates were of the same dilution as the DMSO.
The dilutions of the extract samples are shown in Table 6 below and those for IFNα are shown in Table 7.
Table 6.
Figure imgf000033_0002
Table 7.
Figure imgf000033_0003
Figure imgf000034_0001
In the first assay the replicon cells were plated out at a concentration of 5 x 103 / well in 90μl. The following day the test samples were added in lOμl. The plates were incubated for a further 72 hours and harvested and assayed using the Dual Luciferase Assay Promega. The replicon cell line expresses Renilla luciferase.
The samples were tested for cytotoxicity by setting up a separate 96 well plate with each sample and concentration in duplicate. The cells were labeled with tritiated thymidine for 24 hours and harvested.
For the second assay a separate plate with the top 5 concentrations was set up and incubated for 2 hours with wst-1 (Roche) and the OD read at 450nm and 630nm. Wst-1 is a viable cell stain.
Results
Assay 1: Mean of reading from cells without any drug added 198906. The results are shown in Table 8.
Table 8.
% inhibition of expression of Renilla luciferase
Figure imgf000034_0002
Figure imgf000035_0001
The readings obtained with no added drug were quite high.
The result obtained with the interferon at these concentrations is typical of this assay. It seemed from these results that inhibition was obtained with some of the samples at the higher concentrations so the assay was repeated.
Assay 2.
The results are shown in Table 9
Table 9.
Mean of reading from cells with no drug added. 71676.
% inhibition of expression of Renilla lucif erase
Figure imgf000035_0002
The dilutions were tested for cytotoxicity in the wst-1 assay and this plate was also examined under the microscope. This assay will not detect cytostatic agents. Sample 4 killed the cells at the top 2 dilutions; they looked less healthy at the 3rd concentration. AU the other samples had cells that were healthy in appearance. This is reflected in the wst-1 results.
Assay 2 wst-1 results.
The results are shown in Table 10
Table 10.
Mean OD reading at 450nm
Figure imgf000036_0001
It should be noted that the IFN alpha concentrations are pg/ml. The results are illustrated graphically in Figs 7 and 8.
Fig 7 shows the raw replicon scores of the samples against concentration. ASC-0705 is clearly the best performer and outperforms IFN alpha (standard treatment). It also demonstrates that the best activity is at a concentration above 10ug/ml.
Fig 8 shows activity against concentration. Clearly the optimum concentration is above about 1.36uM EXAMPLE 5
Further purification
The fraction (14 AS-C) was subjected to a chromatographic separation step as illustrated in Fig 9 and as set out below. Briefly this comprised the following steps:
1. 6.2g of the Dichloromethane extract (14) "AS-C" from Astragalus root was separated by silica gel column chromatography and eluted in gradient elution with chloroform, chloroform/methanol (100:5), chloroform/methanol (100:10), chloroform/methanol (100:15), and methanol and 25 fractions obtained which were initially split into six pools (17; 18; 19; 20; 21 and 22);
2. Pooled fractions 1-14 (17) contained no significant chemicals;
3. Fraction 15 (18) had a weight of 241 mg and was further purified twice with Sephadex column chromatography and eluted with methanol to get a fraction (23)
"AS-C-II" which was shown to be active BUT which also exhibited toxicity;
4. Fraction 17 (19) had a weight of 231 mg and was further separated into two fractions by way of silica gel column chromatography eluting with chloroform/methanol (94:6). The two fractions weighed 13mg, fraction (24) "AS- C-III" and a 14mg (25) "AS-C-IV";
5. The 18th fraction (20) was left to stand and 649mg of a white powder precipitated as AS-C-I. This was found to exhibit activity with no toxicity;
6. The 22nd fraction (21) weighing 219mg was separated by Sephadex column and Silica column to obtain an 18 mg fraction (26) "AS-C-V".
The extracts which were tested are identified in Table 11 below:
Table 11
Ref Sample Source Total % content Factorial weight with change from reference to raw material dry raw material
(14) AS-C 6.2g 0.79671%* 125
(20) AS-C -I AS-C 18th 649mg 0.08339% 1,199 fraction eluted
(23) AS-C -II AS-C 15 th 17mg 0.00218% 45,871 fraction eluted
(24) AS-C -III AS-C 17 rth 13mg 0.00167% 59,880 fraction eluted
(25) AS-C -IV AS-C 17 .th 14mg 0.00179% 55,865 fraction eluted
(26) AS-C -V AS-C 22: nd 18mg 0.00231% 43,290 fraction eluted
* Calculated on basis that lOOg of primary extract (8) contains 12.85% of the starting material and accordingly the equivalent weight of dry raw material is 778.2g
Each of the five referenced sub fraction samples was subjected to analysis by TLC and the results are illustrated in Figs 10 and 11.
Fig 10 shows silica gel plates developed in chloroform / methanol (9:1). The left hand plates is viewed under UV at 254nm and the right hand plate under daylight (spray with 10% sulphuric acid in ethanol and then heat the plate until the spots are clearly seen).
Fig 11 shows silica gel plates developed in chloroform / methanol/ water (8:2:0.2). The left hand plates is viewed under UV at 254nm and the right hand plate under daylight (spray with 10% sulphuric acid in ethanol and then heat the plate until the spots are clearly seen).
In both cases the samples read from left to right: AS-C-I; AS-C-II; AS-C-III; AS-C-IV;
AS-C-V. EXAMPLE 6
Activity
Each of six samples: "AS-C"; "AS-C -I"; "AS-C -II"; "AS-C -III"; "AS-C -IV"; and "AS-C -V" were dissolved in DMSO to give a 10 mg/ml stock solution. The samples were:
1. analysed for purity and identity; and
2. tested for anti HCV activity.
1. Purity and Identity
The six samples, AS-C, AS-C-I5 AS-C-II, AS-C-III, AS-C-IV, AS-C-V were analysed with respect to purity and identity using LC-UV and LC-MS. The analysis gave the following data
• AS-C fraction contained several (>6) small peaks (m/z 269, 301, 303, etc.)
• AS-C-I two main peaks (m/z 269, 301)
• AS-C-II one peak (m/z 301)
• AS-C-III one peak (m/z 303)
• AS-C-IV one peak (m/z 269)
• AS-C-V no peaks As at this stage, the components corresponding to the peaks had not been identified so the only estimation that could be done was to compare the peak areas with the DMSO peak area:
I. "AS-C-II" and "AS-C-IV" contained a single peak which is higher than the DMSO peaks.
II. "AS-C" and "AS-C-I" are mixtures;
III. "AS-C-III" contains a single peak which is lower than the DMSO peak.
IV. "AS-C-V" does not contain any peak visible in UV.
It should be noted that the purity was analysed with UV detection and any compounds that do not absorb in UV will not be detected. Material from the peaks was analysed with MS to determine the m/z (given above).
2. Anti-HCV Activity
The samples were tested for anti-HCV activity in: a) a replicon assay (Reblikon), b) a NS5B assay and c) a NS3/NS4A full length protease assay.
To estimate the selectivity of any "hit" in the replicon assay, the samples were also tested for cytotoxicity in Huh7 cells.
The results are illustrated in Table 12 below which additionally includes purity, peak area and identity data.
Table 12
FRACTION AS-C AS-C-I AS-C-Π AS-C-m AS-C-IV AS-C-V
TEST a) REPLICON, EC50 3.7 36 17 >50 >50 >50 (μg/ml)
b) NS5B, EC50 (μg/ml) 17 19 >100
cl) NS3/NS4, %inh @100 μg/ml 45 46 c2) NS3/NS4, %inh @10 μg/ml 19 27 37
d) Huh7, CCSO (μg/ml) >100 >100 >100
no peaks
Purity (LC-UV) 97% 98% detected
2339866 11689463
PeαA area (2.67 min) (2.57 min)
Identity (LC-MS) m/z 303 m/z 269
Figure imgf000041_0001
From the table the following can be determined:
Sample, "AS-C" (mixture)
• a) Inhibits HCV replication in replicon cells showing EC50 = 3.7 μg/ml;
• b) Inhibits NS5B at 9.2 μg/ml; m
• c) Gives 95 % inhibition at a concentration of 100 μg/ml in the NS3/NS4 assay.
• d) Shows no cytotoxicity in concentrations of up to 100 μg/ml.
AS-C-I and AS-C-II • a) also show some activity in the replicon assay but at higher concentrations. The determined EC50 values were 36 and 17 μg/ml respectively
EXAMPLE 7
Further characterisation
In order to further characterise the "AS-C" fraction (14) which exhibited the strongest activity, reference was made to fraction "AS-C-I" (20) as this also exhibited activity and thus it was concluded it contained one or more of the actives responsible for the activity which could serve as markers.
Accordingly, and with reference to Fig 12, the following protocol was followed:
1. 6.2g of (14) AS-C was separated by silica gel column chromatography and eluted with chloroform/methanol (100:15). The precipitated white powder from the 18th fraction (20) was referred as "AS-C-I" (649mg).
2. The ASC-I (20) was dissolved in methanol with supersonication, and then filtrated. The filtrate was separated by Sephadex LH-20 column to obtain three major compounds as showed in the TLC plates illustrated in Figs 13-15.
3. Pooled fractions 4-6 (28) showed a spot when sprayed with sulphuric acid and heated;
4. Pooled fractions 9-11 (30) showed 2 spots when the plate was placed in an iodine steam and observed at UV254 nm-
5. Pooled fractions 4-6 (28) had trace amounts of pigment after purifying by Silica gel column and eluted with chloroform/methanol: 9:1 (32)
6. This sample (32) was dissolved in acetone, refrigerated and re-crystallized to obtain "AS-C-1-1" (33).
7. Pooled fractions 9-11 (30) showed fluorescent spots when purified by a gel column (34), and then TLC method (35). The two fluorescent bands were separated. The band with a higher Rf value was eluted to obtain "AS-C-I-2" (36); while the one with a lower Rf value was purified by Sephadex LH-20 to obtain "AS-C-I-3" (37).
The three compounds identified were analyzed by mass spectrum and nuclear magnetic resonance spectroscopy, respectively. The structures of the three compounds were identified by comparing the data obtained with the existing literature on chemical structures and are shown in Table 13.
Table 13
Sample No. Spectra tested Structural analysing results
AS-C-I-I ESI, 1HNMR, Astragaloside I 13CNMR, COSY, HMQC, HMBC
AS-C-l-2 ESI, 1HNMR, Formononetin-7-O-β-D- 13CNMR, COSY, glucoside HMQC, HMBC
AS-C-I-3 ESI, 1HNMR, 3'-hydroxyl- HMQC, HMBC formononetin -7-0-β-D- glucoside
These compounds have the following structures:
Formula 1 Astragaloside I (AS-C-I-I)
Figure imgf000044_0001
Formula 2: Formononetin-7-O-β-D-glucoside (AS-C-I-2)
glcO
Figure imgf000044_0002
Formula 3: 3'-hydroxyl- formononetin -7-O-β-D-glucoside (AS-C-I-3)
Figure imgf000044_0003
EXAMPLE 8
Investigation of activity against Dengue Virus
The objective was to test: a) The cytotoxicity of extracts ASC, ASC 90(30) and ASC 90(5O)5 and b) Test the effects of extracts ASC, ASC 90(30) and ASC 90(50) on dengue virus infection METHOD
Preparation of test solutions
Each test sample (ASC 0506, ASC 90 (30) and ASC 90 (50) (10 mg) was re-suspended in 1 ml of DMSO to obtain the concentration of 10 mg/ml. The completely dissolved extract was diluted by a 100-fold with growth medium and, therefore, this stock solution had the final concentration of 100 μg/ml (in the presence of 1% DMSO). To test the effects of the extract on cell viability and dengue virus infection, the stock solution was serially diluted with the maintenance medium as follows.
100 μg/ml (DMSO = 1%) a 2-fold dilution = 50 μg/ml (DMSO = 0.5%)
Serial 10-fold dilutions: 5 μg/ml (DMSO = 0.05%)
0.5 μg/ml (DMSO = 0.005%)
100 μg/ml (DMSO = 1%) a 5-fold dilution = 20 μg/ml (DMSO = 0.2%)
Serial 10-fold dilutions: 2 μg/ml (DMSO = 0.02%)
0.2 μg/ml (DMSO = 0.002%)
Figure imgf000045_0001
Determination of cytotoxicity
Vero (an African green monkey kidney cell line) or Eahy 926 (an endothelial cell line) were seeded onto 96-well plates (20,000 cells/ well) in 100 μl of growth medium (10% FBS-MEM + 2 niM L-glutamine for Vero cells and 10% FBS-DMEM F-12 for Eahy 926) and incubated at 370C with 5% CO2 for 24 hr. Thereafter, the medium was discarded and the cells were incubated with 100 μl of extracts ASC, ASC 90(30), or ASC 90(50) at the concentrations of 0.2, 0.5, 2, 5, 20, 50, and 100 μg/μl. Cells that had been incubated with 100 μl of the maintenance medium (2% FBSMEM + 2 BiM L-glutamine for Vero cells and 5% FSB-DMEM F-12 for Eahy 926) or DMSO (the diluent of the compounds) at the concentrations of 0.02%, 0.05%, 0.2%, 0.5%, and 1% served as the control. At 24, 48, and 72 hr post incubation, the cells were subjected to an MTT cell proliferation assay (ATCC) according to the manufacturer's protocol. Standard controls for cell proliferation at the indicated time points were also established by plating different numbers of cells in each well:
Figure imgf000046_0001
104, 105, 106 cells/well for Vero or 103, 104, 5 x 104, 105 cells/well for Eahy 926. The absotbance which reflects the number of cells was read out at 595 run (with the reference wavelength of 630 nm). The numbers of DMSO or compound-treated cells were obtained from the predicted value on the standard curve [OD (standard - blank) vs. the number of cells]. The percentage of cell viability was calculated based on the following formula:
% viability = The number of DMSO or compound-treated cells x 100
The number of non-treated cells, all standard controls, DMSO or extract-treated cells, and non-treated cells were tested in triplicate for each experiment. Two independent experiments were performed.
Determination of the effects of the extracts on dengue vivus infection
Vero or Eahy 926 cells were seeded onto 24-well plates (250,000 cells/well) in 1 ml of growth medium (10% FBS-MEM + 2 mM L-glutamine) and incubated at 370C with 5% CO2 for 24 hr. Thereafter, the medium was discarded and cells were incubated with dengue virus serotype 2 (strain 16681) at an MOI of 1 or 5 in the total volume of 250 μl at 37°C for 2 hr. The virus supernatant was then removed and the cells were washed twice with 1 ml of plain medium and further incubated with 1.25 ml of maintenance medium (2% FBS-MEM + 2 mM L-glutamine) which contains compounds ASC, ASC 90(30) or ASC 90(50) at the concentration of 5 or 50 μg/μl, or DMSO (the diluent of the compounds) at the concentration of 0.05% or 0.5%. At 24, 48, and 72 hr post infection, mock (negative control) and dengue virus infected cells were harvested and assayed for the viability by staining with tryptan blue or propidium iodide. The percentage of dengue virus infection was assessed by immunofluorescence staining of intracellular dengue viral E and NSl and subsequent analysis by flow cytometry. In addition, titers of infectious virus in the culture supernatants were determined at the indicated time points by a focus forming unit assay.
RESULTS Cytotoxicity
Vero and Eahy 926 cells were cultured in the presence of extracts ASC, ASC 90(30), and ASC 90(50) at the concentrations of 0.2, 0.5, 2, 5, 20, 50, and 100 μg/ml up to 3 days. The extract-treated cells were harvested daily and determined for the viability by an MTT assay based on cell proliferation. DMSO-treated cells collected at the same time points served as the control. Results from two independent experiments showed that all extracts did not cause significantly cell death in both cell lines tested but, instead, were likely to induce cell proliferation as compared with the control. In addition, cell viability appeared to be time-dependent since it became decreased on day 3 after culture establishment. Both the cell lines that had been treated with the extracts still exhibited more than 50% of cell viability even after the highest concentration (i.e., 100 μg/ml) of the extracts was utilized (except on day 3). These results suggested that the cytotoxic concentration (CC50) of each extract may be higher than 100 μg/ml. Therefore, the concentrations of 5 and 50 μg/ml were chosen as a representative of low and high doses of the compounds, respectively, for use in further study on the effects of the extracts on dengue virus infection.
The results are shown graphically in Fig 21.
Activity against Dengue virus infection
Vero and Eahy 926 cells were incubated with dengue virus serotype 2 (strain 16681) at an MOI of 1 or 5 for 2 hr and thereafter the virus supernatant was replaced with fresh medium containing extracts ASC, ASC 90(30), or ASC 90(50) at the final concentrations of 5 or 50 μg/ml. The cultures were further incubated up to 3 days. Mock and dengue virus-infected cells that had been treated or left untreated with DMSO served as controls for dengue virus infection.
In dengue virus-infected Vero cell culture (MOI=I and 5), treatment with extracts at the concentration of 5 μg/ml yielded the similar percentage of viable cells as compared with DMSO-treated control. The viability of dengue virus-infected cells tended to decrease in the presence of 50 μg/ml of the extracts, particularly ASC and ACS 90(50), as determined by tryptan blue and propidium iodide staining. Dengue virus infection resulted in the expression of viral E and NSl antigens in Vero cells in a dose-dependent and a time-dependent manner (Fig 22) and almost all of the cells (about 90%) could be infected with dengue virus by day 3 post infection. Regardless of the virus doses, treatment with the extracts at 5 μg/ml did not affect the percentage of viral antigen expression as well as the production of infectious virus as assessed by immunofluorescence staining and a focus forming unit assay, respectively. However, treatment with ASC and ASC 90(50), but not ASC 90(30), at 50 μg/ml (Fig 23) resulted in a lower level of virus production by about one log10 FFU/ml, than treatment with DMSO in the dengue virus-infected cell culture (MOI=5). When Vero cells were infected with a lower dose of virus (MOI=I), a slightly reduction of viral antigen expression was observed following treatment with 50 μg/ml of either the ASC or the ASC 90(50). More interestingly, the supernatants of these culture had a significant decrease in the virus titer by about 2-3 log10 FFU/ml as compared with that of the DMSO-treated control. Dengue virus-infected Eahy 926 cell culture (MOI=5) showed the profile of cell viability upon treatment with the extracts, similar to dengue virus-infected Vero cell culture, at both the concentrations tested. Specifically, treatment with 50 μg/ml of the ASC and ASC 90(50) as well as, to a lesser extent, the ASC 90(30), tended to induce cell death in the infected Eahy 926 culture at a higher level than DMSO-treated control. Following dengue virus infection, relatively low levels of viral E and NSl expression were detected regardless of treatment with the extracts; therefore, it would be somewhat difficult to assess the extract activity against dengue virus infection based on the viral antigen expression under the condition tested. A focus forming unit assay was utilized as an alternative approach to determine the effect of the extracts on dengue virus production. Results showed that, virus titers in dengue virus-infected Eahy 926 cell culture containing 50 μg/ml of the ASC and ASC 90(50) were about 2-3 log10 FFU/ml lower than those in the DMSO- treated or the ASC 90(30)-treated cultures. This was consistent with the results observed in dengue virus-infected Vero cell culture
Conclusion
Extracts ASC, ASC 90(50) and ASC 90(30) at all the concentrated tested were not toxic to Vero and Eahy 926 cells in the absence of dengue virus infection. The predicted CC50 of each extract may therefore be higher than 100 μg/ml. However, treatment with 50 μg/ml of the ASC and ASC 90(50) resulted in certain extents of cell death in dengue virus-infected Vero and Eahy 926 cultures. This phenomenon was coincident with a slightly reduction of viral antigen expression and a significant decrease in virus production in the same cultures following treatment with these extracts. The induction of cell death by the extracts ASC and ASC 90(50) may be one of the potential mechanisms to reduce the initial pool of infectious viruses during dengue virus infection of the susceptible cells.
The benefits of the reduction in viral load can be most effectively demonstrated with reference to Figs 19 and 20. Thus, where a patient presents with symptoms (Fig 19) early use of the extract can result in a reduction of viral load on a log scale with the benefit (Fig 20) that the patient develops a "fever" as opposed to a "hemorrhagic fever".

Claims

1. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament for the treatment of a disease caused by a genus of the flaviviridae family selected from:
• the genus - Flavivirus, (Table 1.1);
• the genus - Pestivirus, (Table 1.2);
• the genus — Unassigned Flaviviradae (Table 1.3) or
• tentative Species in the Genus HCV (Table 1.4).
2. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 1 in which the Flavivirus is a mosquito bourne flavivirus.
3. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 2 wherein the mosquito bourne flavivirus is selected from: o Dengue Virus (DEN); o West Nile (WN) virus; o Japanese Encephalitis (JE) virus; o Kunjin virus (KUN); o Murray Valley encephalitis virus (MVEV); o St. Louis encephalitis virus (SLEV); and o Yellow fever.
4. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 1 in which the Flavivirus is a tick bourne flavivirus.
5. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 4 wherein the tick bourne flavivirus is selected from: o Kyasanur Forest disease virus o Langat virus o Louping ill virus o Omsk hemorrhagic fever virus o Powassan virus o Tick-borne encephalitis virus
6. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 1 in which the Pestivirus is selected from:
• Bovine viral diarrhea virus 2, and
• Classical swine fever virus.
7. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 1 in which the unassigned Flaviviridae is selected from:
• GBV-A5 and
• GBV-C.
8. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in claim 1 in which the tentative Species in the Genus Hepacivirus, is o GBV-B
9. An Astragalus extract or an active fraction thereof or an active compound isolated therefrom, for use in the manufacture of a medicament as claimed in any of the preceding claims in which the extract, active fraction thereof or compound isolated therefrom is or is derived from a single herb extract.
10. An Astragalus extract, or an active fraction thereof or compound isolated therefrom as claimed in any of the preceding claims wherein the active fraction is an extract of Astragalus purified by at least a factor of 10 with reference to dried raw plant material, and which is characterized in that it comprises at least one marker selected from the group consisting of:
• Astragaloside I;
• Astragaloside IV
• Formononetin-7-o-β-d-glucoside; and
• 3 ' -hydroxyl-formononetin-7-o-β-d-glucoside.
11. An Astragalus extract, or an active fraction thereof or compound isolated therefrom as claimed in claim 10 wherein the active fraction is an extract of Astragalus purified by a factor of 50 with reference to dried raw plant material.
12. An Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in claim 11 wherein the active fraction is an extract of Astragalus purified by a factor of between 75 and 200 with reference to dried raw plant material.
13. An Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in any of claims 9-12 which is derived from an alcoholic extract.
14. An Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in claim 13 in which the extract has been subject to an ethanol-water precipitation process.
15. An Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in claim 13 or 14 in which the extract has been subject to a systematic solvent fractionation step with a plurality of solvents of different polarity or separation on a macro-porous resin
16. Pea. Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in claim 15 wherein the active fraction is obtained from a dichloromethane fractionation or a solvent of similar polarity.
17. Pea. Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in claim 15 wherein the extract is separated on a macro- porous resin, washed with water, washed with either 30% or 50% ethanol and eluted with 90% ethanol.
18. An Astragalus extract, or an active fraction thereof or compound isolated therefrom, as claimed in any of the preceding claims wherein the Astragalus is Astragalus membranaceus (Fisch) Bge. or Astragalus membranaceus Bge var. Mongholicus Hsiao.
19. Pea. Astragalus raw material packaged or otherwise sold in a manner which indicates it is suitable and intended for use as an antiviral to treat infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
20. The use of Astragalus or an extract thereof or an active fraction thereof or an isolated compound therefrom in the manufacture of a food, dietary supplement or food additive for use as an antiviral to treat infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4).
21. A method of treating infections caused by a virus from one of: o the genus - Flavivirus, (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4) comprising administering an effective amount of an Astragalus extract or an active fraction thereof or an active compound isolated therefrom to a patient or animal.
22. A method as claimed in claim 21 wherein the Astragalus extract, or an active fraction thereof, is provided in a unit dosage form.
23. A method as claimed in claim 22 wherein the unit dosage form is for oral delivery.
24. A method as claimed in any of claims 21-23 wherein a daily dosage is equivalent to 9-3Og of raw material.
25. A method as claimed in any of claims 23-24 wherein administration is with at least one other immuno-modulatory or anti viral drug.
26. A method as claimed in claim 25 wherein the other drug is interferon and / or Ribavirin.
27. An Astragalus extract fraction having an HPLC fingerprint showing a primary peak for Astragaloside I substantially as shown in any of Figs 16a to 16d.
28. An Astragalus extract fraction as claimed in claim 27 showing a secondary peak for Formononetin-7-O-β-D-glucoside
29. An Astragalus extract fraction as claimed in claim 27 or 28 showing a tertiary peak for Astragaloside IV
30. An Astragalus extract fraction as claimed in any of claims 27-29 comprising no more than seven additional peaks of prominence between the peak for Astragaloside IV and the peak for Astragaloside 1.
31. An Astragalus extract having a TLC fingerprint comprising six identifiable spots substantially as illustrated in any of lanes 2 to 5 of Fig 14 or 15.
32. A method for producing an extract of Astragalus which exhibits antiviral activity against a virus from one of: o the genus - Flavivirus (Table 1.1); o the genus - Pestivirus, (Table 1.2); o the genus - Unassigned Flaviviradae (Table 1.3) or o tentative Species in the Genus HCV (Table 1.4). and which is purified by at least a factor of 10 with reference to dried raw plant material, and which is characterized in that it comprises at least one marker selected from the group consisting of: o Astragaloside I; o Astragaloside IV o Formononetin-7-o-β-d-glucoside; and o 3 ' -hydroxyl-formononetin-7-o-β-d-glucoside comprising:
1) an alcoholic extraction (which may be repeated);
2) an ethanol-water precipitation process (which may be repeated); and either
3.1) a systematic solvent fractionation step with a plurality of solvents of different polarity, or
3.2) a macro-porous resin column purification.
33. A method for producing an extract of Astragalus as claimed in claim 32 wherein the extract is separated on a macro-porous resin, washed with water, washed with a low concentration ethanol and eluted with a high concentration ethanol.
34. A method for producing an extract of Astragalus as claimed in claim 33 wherein the low concentration ethanol is either 35% or 50% ethanol and the high concentration ethanol is 90% ethanol.
PCT/GB2008/003947 2007-11-27 2008-11-26 Antiviral WO2009068872A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0723609.4 2007-11-27
GB0723609A GB2455151A (en) 2007-11-27 2007-11-27 An Astragalus extract as an antiviral for several genera of the Flaviviridae family

Publications (1)

Publication Number Publication Date
WO2009068872A1 true WO2009068872A1 (en) 2009-06-04

Family

ID=38962518

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2008/003947 WO2009068872A1 (en) 2007-11-27 2008-11-26 Antiviral

Country Status (2)

Country Link
GB (1) GB2455151A (en)
WO (1) WO2009068872A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103073614A (en) * 2013-01-22 2013-05-01 西安岳达植物科技有限公司 Method for extracting high-purity astragaloside from astragalus mongholicus
CN104502518A (en) * 2014-12-04 2015-04-08 吉林敖东延边药业股份有限公司 Detection method of traditional Chinese medicine preparation for treating child anorexia

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103808840B (en) * 2012-11-02 2015-09-09 丽珠集团利民制药厂 A kind of method for building up of ginseng and astragalus injection for strengthening body finger-print

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1207911A (en) * 1997-08-08 1999-02-17 梁广斌 Antiviral veterinary medicine injection and its preparation
WO2002047703A2 (en) * 2000-12-11 2002-06-20 Nutricia N.V. Immune system stimulation
CN1607003A (en) * 2003-10-16 2005-04-20 泰宗生物科技股份有限公司 Auxiliary therapeutic agent for C type hepatitis
WO2007020382A2 (en) * 2005-08-12 2007-02-22 Phynova Limited Plant-based medicament for the treatment of liver disease
WO2007144569A2 (en) * 2006-06-16 2007-12-21 Phynova Limited Antiviral astragalus extract

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003504327A (en) * 1999-07-08 2003-02-04 プレンダーガスト、パトリック、ティ. Use of flavones, coumarins and related compounds for the treatment of infectious diseases
JP2007537131A (en) * 2003-06-20 2007-12-20 ヴァイラル・ジェノミックス・インコーポレーテッド Antiviral compositions and methods of using the same
CN1481805A (en) * 2003-07-14 2004-03-17 军 王 Medication for hepatitis and its preparation method
CN1985877A (en) * 2006-12-30 2007-06-27 中国人民解放军总医院 Astragalus extract and its medicine use

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1207911A (en) * 1997-08-08 1999-02-17 梁广斌 Antiviral veterinary medicine injection and its preparation
WO2002047703A2 (en) * 2000-12-11 2002-06-20 Nutricia N.V. Immune system stimulation
CN1607003A (en) * 2003-10-16 2005-04-20 泰宗生物科技股份有限公司 Auxiliary therapeutic agent for C type hepatitis
WO2007020382A2 (en) * 2005-08-12 2007-02-22 Phynova Limited Plant-based medicament for the treatment of liver disease
WO2007144569A2 (en) * 2006-06-16 2007-12-21 Phynova Limited Antiviral astragalus extract

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Week 199926, Derwent World Patents Index; AN 1999-303278, XP002526292 *
DATABASE WPI Week 200559, Derwent World Patents Index; AN 2005-572039, XP002482221 *
RIOS J L ET AL: "A review of the pharmacology and toxicology of Astragalus", 1 September 1997, PHYTOTHERAPY RESEARCH, JOHN WILEY & SONS LTD. CHICHESTER, GB, PAGE(S) 411 - 418, ISSN: 0951-418X, XP002475583 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103073614A (en) * 2013-01-22 2013-05-01 西安岳达植物科技有限公司 Method for extracting high-purity astragaloside from astragalus mongholicus
CN103073614B (en) * 2013-01-22 2016-04-20 西安岳达植物科技有限公司 A kind of method extracting Cyclosiversioside F from the Radix Astragali
CN104502518A (en) * 2014-12-04 2015-04-08 吉林敖东延边药业股份有限公司 Detection method of traditional Chinese medicine preparation for treating child anorexia

Also Published As

Publication number Publication date
GB0723609D0 (en) 2008-01-09
GB2455151A (en) 2009-06-03

Similar Documents

Publication Publication Date Title
Manvar et al. Identification and evaluation of anti hepatitis C virus phytochemicals from Eclipta alba
Naithani et al. Antiviral activity of phytochemicals: a comprehensive review
Li et al. Anti-hepatitis B virus activities of Geranium carolinianum L. extracts and identification of the active components
Suzuki et al. Anti-hepatitis C virus effect of citrus unshiu peel and its active ingredient nobiletin
Wang et al. Substituting one Paris for another? In vitro cytotoxic and in vivo antitumor activities of Paris forrestii, a substitute of Paris polyphylla var. yunnanensis
He et al. The NCI library of traditional Chinese medicinal plant extracts–Preliminary assessment of the NCI-60 activity and chemical profiling of selected species
CN106420902A (en) Hepatoprotective activity and new medical application of glycyrrhiza inflata extract and licochalcone A
CN104370871B (en) The mouth diphenylene ketone oxide class separated from Swertia punicea Hemsl. and the application of suppression hepatitis B virus
Samy et al. Phytochemical investigation of Amphilophium paniculatum; an underexplored Bignoniaceae species as a source of SARS-CoV-2 Mpro inhibitory metabolites: Isolation, identification, and molecular docking study
US20120269768A1 (en) Antiviral product
Naithani et al. Antiviral activity of phytochemicals: a current perspective
WO2009068872A1 (en) Antiviral
Bai et al. Advances in the chemical constituents, pharmacological properties and clinical applications of TCM formula Yupingfeng San
Elkousy et al. Anti-SARS-CoV-2 in vitro potential of castor oil plant (Ricinus communis) leaf extract: in-silico virtual evidence
Song et al. Flavonoids from the seeds of hovenia acerba and their in vitro antiviral activity
Mandal et al. Medicinal plant molecules against hepatitis C virus: Current status and future prospect
JP2000503686A (en) Pharmaceutical composition for the treatment of hepatitis C, comprising a mixed extract of yellow bamboo skin and Ominaeushi plant
US20160213730A1 (en) Identification of natural plant extracts harboring anti-hepatitis c virus ns5b polymerase activity
KR100704006B1 (en) Anti-cancer activity of Androsace umbellata Merr. extract and contained triterpene saponin
Lin et al. Comprehensive LC-MS/MS-based phytochemical perspectives and osteogenic effects of Uraria crinita
Sivagami et al. A review on analytical methods for antiviral Phytoconstituents
CN114957190A (en) Artemisia japonica extract, pharmaceutical composition thereof, preparation method and application thereof
Gowtham et al. Exploring structurally diverse plant secondary metabolites as a potential source of drug targeting different molecular mechanisms of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) pathogenesis: An in silico approach
Séron et al. Natural products and hepatitis C virus
Apriyanto et al. Potential of Indonesian plants (Annona muricata, Garcinia latissima, and Garcinia celebica) against hepatitis C virus

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08853325

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08853325

Country of ref document: EP

Kind code of ref document: A1