WO2000001398A1 - Extrait de plante antiviral - Google Patents

Extrait de plante antiviral Download PDF

Info

Publication number
WO2000001398A1
WO2000001398A1 PCT/IB1999/001235 IB9901235W WO0001398A1 WO 2000001398 A1 WO2000001398 A1 WO 2000001398A1 IB 9901235 W IB9901235 W IB 9901235W WO 0001398 A1 WO0001398 A1 WO 0001398A1
Authority
WO
WIPO (PCT)
Prior art keywords
plant
extract
patient
hiv
powder
Prior art date
Application number
PCT/IB1999/001235
Other languages
English (en)
Inventor
Nobutho Gladys Makupula
Original Assignee
Nobutho Gladys Makupula
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 Nobutho Gladys Makupula filed Critical Nobutho Gladys Makupula
Priority to AU45275/99A priority Critical patent/AU4527599A/en
Publication of WO2000001398A1 publication Critical patent/WO2000001398A1/fr

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)
    • A61K36/47Euphorbiaceae (Spurge family), e.g. Ricinus (castorbean)
    • 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
    • A61P31/18Antivirals for RNA viruses for HIV

Definitions

  • This invention relates to a composition or substance for use in the treatment of diseases affecting the human immune system and provides for methods of treatment using it and for preparing it.
  • AIDS Acquired immunodeficiency syndrome
  • HIV human immunodeficiency virus
  • HIV is thought to contribute to the causing AIDS by infecting helper/inducer T4 cells in a patient's body. This leads to a T4 cell deficiency, opening the way for further infection.
  • Japanese patent publication number 7-165600 discloses an antiviral agent having anti-HIV action, such agent containing an extract of Euphorbia kansui Liou and a terpene derivative prepared from this species or from Euphorbia Kansui radix.
  • a suite of United States patents issued to Herman disclose AIDS and HIV remedies that comprise substances that may be derived from plant sources, in particular various terpenes and terpene derivatives, among them lupeol. Particular reference may be made to Herman's US5260342, disclosing the parenteral application of a pharmacologically antivirally effective amount of an ozonide of a terpene to fight AIDS and other viral infections in mammals. Further reference may be had to US5126376, US5190979 and US4983637.
  • a composition for use in a method of treatment of an immune system disorder in the human body comprises an antiviral agent effective against HIV, administrable to a patient in a therapeutically effective amount and derived from a plant of the species Euphorbia gorgonis.
  • the agent is derived from a root portion of the plant, such root portion including a bulb portion.
  • composition may be provided for administration in solid or liquid form. Administration may be accomplished orally or parenterally.
  • the liquid of the composition is the decantate or filtrate of a mixture of particulate matter derived from the plant and a liquid carrier.
  • the invention thus includes pharmaceutical compositions for use in the treatment of HIV infection comprise an extract from a plant of the species Euphorbia gorgonis, such extract being in a pharmacologically antiviral dosage amount.
  • the dosage amount is in the range of 4 to 12mg of dried extract powder per kilogram of patient body mass daily and is administered in two or more equal doses. Further preferably, the range is from 6 to 9 mg/kg of patient body mass daily.
  • the invention extends to the use of an Euphorbia gorgonis plant in providing an extract for the treatment of HIV when administered in a therapeutically effective amount to a patient. Administration is preferably by oral means.
  • a method of treating HIV infection in a human patient includes introducing to such patient an extract derived from a plant of the species Euphorbia gorgonis in an antivirally effective dose and repeating the dose daily.
  • the total daily dose is administered at least twice per day in substantially equal portions.
  • each dose comprises a dried powdered extract from a root portion of the plant.
  • the total daily dose of powder is preferably in the range of from 4mg/kg to 12mg/kg of body mass of the patient.
  • the powder dose may be dissolved in a liquid carrier.
  • a preferred concentration range is from 5 to 10 grams per litre of carrier.
  • the invention extends to a method of manufacturing an antiviral composition effective in treating a patient infected with HIV, the method including the steps of providing plant material from a plant of the species Euphorbia gorgonis, producing an extract from the plant material that includes an antiviral agent effective against HIV in a form suitable for administration to such patient.
  • the extract is derived from a root portion of the plant, such portion including a bulb of the plant.
  • the extract is formulated to be available in therapeutically effective dosage amounts.
  • Figures 1 and 2 show results of cytotoxicity tests on CEM-SS cells exposed to extracts of Euphorbia gorgonis as described in Example 1.
  • Figure 3 shows the effect of treatment with sample extracts S1 through S4 of Example 1 on the production of p24 antigen in HIV-1 infected cultures as measured by optical absorbance at 450nm.
  • Figure 4 is a graph showing the effect of extract S5 of Example 1 on the production of p24 antigen in HIV-1 infected cultures expressed as a percentage of the absorbance of untreated virus-infected cultures.
  • Figure 5 is a bar graph showing the advantageous effect of extract S5 of Example 1 on the amount of p24 antigen produced in treated HIV-1 infected cultures.
  • Figure 6 is a graph showing the effect of extract S6 of Example 1 on production of p24 antigen in treated HIV-1 infected cultures as reflected by absorbance at 450nm.
  • Figure 7 provides a set of line graphs on common axes illustrating the results of clinical trials wherein the composition of the invention was administered to HIV infected patients in examples 3 and 4.
  • the plants suitable for use in preparing the composition of the invention are members of the species Euphorbia gorgonis. These plants have a root portion comprising a relatively large bulb, a large specimen being about the size of an adult person's fist. Hair-like root fibres extend from the bulb. The bulb portion is the preferred part of the plant for processing and thereby extracting antiviral agent in the composition.
  • the plant extract, in which the antiviral agent is isolated is, in preferred embodiments of the invention, processed into a pharmaceutical composition for use in a method of treating a patient infected with HIV or AIDS.
  • a pharmaceutical composition for use in a method of treating a patient infected with HIV or AIDS.
  • Such composition may include more than one active ingredient. Analysis has indicated the presence of lupeol derivatives in the extract.
  • Pharmacologically antiviral dosage amounts may then be made up for easy administration to a patient.
  • Methods of preparation include extraction by alcohol or water.
  • a preferred alcohol is ethanol, although others may be used, depending in compatibility with the active ingredients and toxicity to the patient.
  • Specific examples of methods for preparing a composition suitable for administration to humans are described in Examples 2 and 3 below. These methods broadly include the steps of providing dried plant material, selecting the bulb portion, dividing it into small portions for further drying, for example by slicing, reducing the dried material into powder form, and dissolving the powder in a liquid. The dissolving step is preferably accompanied by heating. The solution is preferably thereafter left to cool and the supernatant liquid recovered.
  • dissolving a mass of between 5g and 10g of dried powder per litre of solvent provides a therapeutically effective composition for reducing HIV-1 infection in adult patients who had been administered a daily dose of 60ml over a period of at least about 50 days.
  • a greater concentration than 10g/l of powder may be required to be effective, or alternatively, more that 60ml of the therapeutic solution.
  • composition may be administered in either solid or liquid form, so as to be received by a patient orally, by injection, absorption, inhalation or other method well known in the art. It is therefore within the scope of this invention that the composition may be suitably formulated to be included in the form of nose- and ear-drops, nasal and throat sprays, ointments, creams and suppositories.
  • the plant material obtained by drying and dividing the root and in particular the bulb portion is preferably compressed into dosage sized particles, convenient for swallowing.
  • compression may be in the presence of a suitable compatible binder material.
  • the particulate solids may also be administered orally in powder form. The powder may be placed under a patient's tongue, or simply swallowed, with or without the aid of a liquid.
  • the powdered particulate matter of the composition may be mixed or dissolved into a suitable liquid carrier and the mother liquor from the mixture decanted or filtered off.
  • the carrier may be water, or alternatively an extract may be prepared with the use of a suitable organic solvent which does not lead to the degrading of pharmacologically active constituents. It has been found that administering about 60ml of the liquid a day to HIV-infected patients for a period of 50 days or more was effective to reduce their HIV-1 cell count significantly.
  • the powder may be obtained by fragmenting the bulb or root portions of the plant and crushing the fragments. Alternatively, these portions may be ground of grated or subjected to other suitable size reduction methods.
  • Dried powder extracts obtained by hexane extraction and subsequently purified, have been found to be effective in reducing the p24 antigen count in CEM-SS cells infected with HIV-1 when administered in concentrations of 20 ⁇ g/ml and above.
  • Preferred concentrations of such extract for p24 antigen reduction are in the range from 20 ⁇ g/ml to 65 ⁇ g/ml.
  • a further preferred concentration range is from 25 ⁇ g/ml to 50 ⁇ g/ml.
  • the solid preferably comprises the powder obtained as referred to above.
  • a patient may receive a therapeutically effective dose of the substance by consuming suitably sized portions of the bulb of the plant in its raw, unprocessed state, either undried or dried.
  • Dosage ranges may vary according to patient condition. However, clinical trials have indicated a preferred dosage range for human patients to be from 4mg to 12 mg of powder per kilogram of patient body mass per day. A further preferred range is from 6 to 9mg/kg body mass. A most preferred range is shown to be from 7-8mg/kg of body mass per day.
  • the daily dosage may be administered in two or more substantially equal portions, for example twice a day in the morning and evening, or three times a day at conventional meal times. It has been found that treatment should be continued for at least 50 days before favourable results in the reduction of infection levels in patients having a HIV-1 viral loading in excess of 250 000 HIV RNA copies/ml plasma are confirmed.
  • composition has potential uses as a broad-spectrum antiviral agent. Apart from its uses in combating HIV and AIDS, the inventive composition has been used with success in the test treatments of other sexually transmitted diseases and cancer. A process for manufacturing the composition of the invention is described in the example that follows. Results of a clinical trial on an AIDS sufferer are also given
  • the T- cell count of a patient receiving the extract is monitored. If the T-cell count increases, this means the patient is responding positively to the treatment. One expects the patient's CD4 cell count to increase correspondingly, these being housed within the T-cells. Where the CD4 count does not improve, the indication is that the virus Is still present.
  • CEM-SS syncitium-sensitive lymphoblastoid line
  • RPMI 1640 medium containing L-glutamine (Whittaker Bioproducts, Md) and supplemented with 10% heat-inactivated fetal bovine serum (Delta Bioproducts, Kempton Park, South Africa).
  • the cells were passaged every second day to maintain a culture concentration of 1 x 10 6 cells/ml.
  • HIV-1 stocks prepared from a subtype D strain (Engelbrecht et al., 1995) and stored at -80°C, were used.
  • the product of the methanol extraction was re- dissolved in 50% DMSO (at room temperature) at a final concentration of 5 mg/ml.
  • the two compounds purified from the hexane extract were re-dissolved in 100% DMSO (with heating over an open flame) at a concentration of 10 mg/ml.
  • the prepared stock solutions were stored at -80°C. Before use the stock solutions were diluted in complete medium to a concentration of 1 mg/ml and filter sterilized through a 0,45 ⁇ m filter.
  • the final DMSO concentration in each of the test assays was not more than 1 % (v/v) (Gustafson et al, 1992).
  • Each extract or compound was tested in quadruplet at concentrations of 5 ⁇ g/ml, 10 ⁇ g/ml, 25 ⁇ g/ml, 50 ⁇ g/ml, 75 ⁇ g/ml and 100 ⁇ g/ml in a 96 well cell culture plate.
  • the CEM-SS cells were seeded at a density of 1x10 5 cells/well and the extracts or pure compounds were added at the required concentrations in a final culture volume of 200 ⁇ l/well.
  • the treated cell cultures were incubated at 37°C in a C0 2 incubator for three days, after which they were observed by light microscopy for any morphological changes indicative of toxicity.
  • the tetrazolium salt (MTT) reduction assay was used to measure cell viability (Mossman et al, 1983).
  • the trypan-blue exclusion method was used to determine the cell concentrations used in the test assays.
  • Each of the 4 extracts and the 2 compounds purified from the hexane extract were tested at three concentrations in quadruplet against a virus concentration of 100 TCID 50 .
  • Untreated infected and uninfected CEM-SS cells were included as controls.
  • the cells at a concentration of 5 x 10 5 cells/ml were pre-incubated with virus for 60 minutes at 37°C. After incubation the cells were washed twice with PBS and the extracts and compounds were added at the required final concentrations in complete medium.
  • the total culture volume was 200 ⁇ l/well.
  • the culture plates were then incubated at 37°C in a C0 2 incubator. At day 4, post- infection SNF from each well was collected for p24 antigen assay.
  • the Vironostika® HIV-1 Microelisa System (Organon Teknika, USA) for qualitative and semi-quantitative detection of the p24 core antigen of HIV-1 , was used to measure p24 antigen in the cell culture SNF. NMR testing
  • the absorbance of the reduced tetrazolium salt is a direct measure of the activity of lactate dehydrogenase (LDH) activity in living cells.
  • LDH lactate dehydrogenase
  • the hexane extract (SI) showed reduced absorbance - and hence cytotoxicity - at extract concentrations of 50 ⁇ g/ml to 100 ⁇ g/ml (figure 1 ).
  • the methylene chloride (S2) and ethyl acetate (S3) extracts were not significantly cytotoxic to the CEM-SS cells at extract concentrations from 5 ⁇ g/ml to 100 ⁇ g/rnl (figure 1 ).
  • the methanol extract (S4) was not cytotoxic to the CEM-SS cells at extract concentrations from 5 ⁇ g/ml to 100 ⁇ g/ml (figure 2).
  • S5 purified from the hexane extract showed reduced absorbance at a concentration of 100 ⁇ g/ml in S5 treated CEM-SS cells (figure 2).
  • S6 also purified from the hexane extract was cytotoxic to the CEM-SS cells at concentrations from 75 ⁇ g/ml to 100 ⁇ g/ml (figure 2). Further, the cells did not look healthy microscopically when treated with S6 at 25 ⁇ g/ml and 50 ⁇ g/ml.
  • the sample S1 powder resuspended in DMSO did not reduce the production of p24 antigen in HIV-1 infected cultures at concentrations of 10 ⁇ g/ml, 25 ⁇ /ml, and 35 ⁇ g/ml when compared to untreated HIV- 1 infected cultures.
  • the resuspended S2 powder did not reduce the production of HIV-1 p24 antigen in treated virus-infected cultures when compared to untreated virus infected cultures.
  • the resuspended S3 powder did not reduce p24 antigen production in HIV-1 infected cultures treated with 25 ⁇ g/ml, 50 ⁇ g/ml, and 75 ⁇ g/ml of S3. Furthermore, there was no reduction of p24 antigen in HIV-1 infected cultures treated with 25 ⁇ g/ml, 50 ⁇ g/ml and 100 ⁇ g/ml of sample S4.
  • the compound S5 purified from the hexane extract, reduced the p24 antigen produced by HIV-1 infected cultures by 5% and 22% respectively at concentrations of 25 ⁇ g/ml and 50 ⁇ g/ml.
  • the control p24 antigen concentrations were plotted against their signal/cutoff values to produce a semi- quantitative calibration curve.
  • the amount of p24 antigen produced in S5-treated cultures was determined by interpolation of the signal/cutoff values on the calibration curve.
  • the S5 treated HIV-1 infected cell cultures produced >160 ⁇ g/ml, 145 ⁇ g/ml and about 75 ⁇ g/ml p24 antigen in the presence of 10 ⁇ g/ml, 25 ⁇ g/ml and 50 ⁇ g/ml of S5, respectively, as illustrated in figure 5.
  • the count of p24 antigen produced in the untreated virus infected cultures and the virus infected cultures treated with 10 ⁇ g/ml S5 was estimated, because it was more than the 160 ⁇ g/ml limit of the calibration curve.
  • the untreated HIV-1 infected cultures produced more than 160 ⁇ g/ml p24 antigen (figure 5).
  • the compound S6, also purified from the hexane extract did not reduce the production of p24 antigen in HIV-1 infected cell cultures treated with 5 ⁇ g/ml, 10 ⁇ g/ml and 25 ⁇ g/ml of S6, as will be seen when referring to figure 6.
  • a concentration of 25 ⁇ g/ml of S6 was also toxic to the cultures as observed microscopically.
  • the amount of p24 antigen produced in the SI, S2, S3, S4, and S6 treated HIV-1 infected cultures was not reduced.
  • the purified compound S6 was the most toxic to the CEM-SS cells at concentrations from 25 ⁇ g/ml and higher.
  • the S1 extract was toxic to the CEM-SS cells at concentrations of 50 ⁇ g/ml to 100 ⁇ g/ml.
  • the S5 compound showed toxic effects on the CEM-SS cells at a concentration of 100 ⁇ g/ml.
  • S5 Only the purified compound, S5, inhibited the replication of HIV-1 in CEM-SS cells as observed by the reduction in p24 antigen production in infected cultures treated with 25 ⁇ g/ml and 50 ⁇ g/ml of S5. At a concentration of 50 ⁇ g/ml, S5 reduced the amount of p24 antigen produced in HIV-1 infected cultures by more than 50%. The compound designated S5 therefore appeared to present the best potential for developing a composition suitable for administering to humans.
  • the powder was added to water and brought to the boil.
  • the proportions used were about 5 tablespoons (or about 30g) powder added to 4 litres of water. After being brought to the boil, the heat source was removed and the mixture allowed to stand and cool to ambient.
  • the liquid was thereafter decanted to provide a liquid medication while the residue was allowed to dry and collected in powder form for a solid medication form.
  • An AIDS patient infected with HIV was treated by administering doses comprising three tablespoons (i.e. 3 x 10ml) of the powder solution prepared according to the example above, twice daily before meals in the morning and evening respectively.
  • the total daily dose was therefore about 60ml of the solution.
  • a therapeutic composition according to the invention was manufactured as follows: A number of healthy Euphorbia gorgonis plant specimens were picked and allowed to dry indoors for two days under ambient conditions, whereafter the leaves and stems were separated from the bulbous root portions. The latter were then sliced in to pieces about 2mm thick and allowed to dry further until in a condition to be grated into powder having a meal-like consistency.
  • An alcohol based extract was then prepared by first providing a solution comprising 18g of table salt (sodium chloride) in 1.6 litres water and mixing into this 400mi of ethanol 90% commercial grade alcohol. A mass of 16g of the grated plant powder was then added to this solution. The solution was heated to a temperature of about 72°C, being just below the boiling temperature of the alcohol and thereafter allowed to cool.
  • table salt sodium chloride
  • This extract as contained in the cooled solution was administered twice a day to an adult male patient, referred to below as "Patient 1" in a clinical trial described below.
  • Immundeficiency screening was performed to determine the patient's infection level by way of a HIV viral load test, according to conventional pathological practice.
  • a HIV viral load test was performed on a blood sample extracted from this male adult patient and yielded a value of 13470 HIV-1 copies per millilitre.
  • the patient was administered the therapeutic solution in twice daily doses of 30 ml. Dosage levels were about 8mg/kg of the patient's body mass per day at the start of the trial.
  • a water based extract was prepared according to the method described in Example 1 above and was strained to remove suspended particles and sediment.
  • the initial HIV loading determined for this 48 year old female patient was 17090 RNA copies per millilitre of her blood plasma. After an initial increase in her HIV loading levels in the two months following commencement of treatment, her loading decreased rapidly and tailed off to below measurable levels of 500 copies/ml plasma. Treatment ceased and her levels were again at intervals. No increase was observed.
  • HIV PCR 535 058 copies/ml
  • T-Cells 195 cells/cmm
  • HIV Loading is expressed in units of RNA copies per millilitre of plasma

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Virology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Botany (AREA)
  • Biotechnology (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Mycology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

L'invention concerne une composition antivirale de traitement d'infection à VIH ou du SIDA et comprenant un extrait dérivé d'une plante de l'espèce Euphorbia Gorgonis et elle s'étend à une composition pharmaceutique comprenant une dose thérapeutique efficace de cet extrait. L'extrait est dérivé de préférence de la partie bulbeuse de la racine de la plante par un procédé d'extraction consistant en la division de la matière de la plante et sa fragmentation puis sa réduction en une forme pulvérulente. La poudre peut être administrée à un patient ou elle peut être dissoute dans un excipient liquide approprié.
PCT/IB1999/001235 1998-07-02 1999-07-02 Extrait de plante antiviral WO2000001398A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU45275/99A AU4527599A (en) 1998-07-02 1999-07-02 Anti-viral plant extract

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ZA98/5796 1998-07-02
ZA985796 1998-07-02

Publications (1)

Publication Number Publication Date
WO2000001398A1 true WO2000001398A1 (fr) 2000-01-13

Family

ID=25587111

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB1999/001235 WO2000001398A1 (fr) 1998-07-02 1999-07-02 Extrait de plante antiviral

Country Status (2)

Country Link
AU (1) AU4527599A (fr)
WO (1) WO2000001398A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1572220B1 (fr) * 2002-12-11 2008-02-13 Achidi Valentin Agon Proprietes antivirales des extraits de dichrostachys glomerata

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07165600A (ja) * 1993-10-18 1995-06-27 Souyaku Gijutsu Kenkyusho:Kk 抗ウイルス剤
WO1996027383A1 (fr) * 1995-03-06 1996-09-12 Alix Roland Commin Compositions possedant des proprietes antivirales et procede d'obtention
JPH09194387A (ja) * 1996-01-22 1997-07-29 Soyaku Gijutsu Kenkyusho:Kk トウダイグサ科植物の有機抽出物を含有する抗hiv剤

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH07165600A (ja) * 1993-10-18 1995-06-27 Souyaku Gijutsu Kenkyusho:Kk 抗ウイルス剤
WO1996027383A1 (fr) * 1995-03-06 1996-09-12 Alix Roland Commin Compositions possedant des proprietes antivirales et procede d'obtention
JPH09194387A (ja) * 1996-01-22 1997-07-29 Soyaku Gijutsu Kenkyusho:Kk トウダイグサ科植物の有機抽出物を含有する抗hiv剤

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch Week 199534, Derwent World Patents Index; Class B02, AN 1995-178540, XP002118722 *
PATENT ABSTRACTS OF JAPAN vol. 1997, no. 11 28 November 1997 (1997-11-28) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1572220B1 (fr) * 2002-12-11 2008-02-13 Achidi Valentin Agon Proprietes antivirales des extraits de dichrostachys glomerata

Also Published As

Publication number Publication date
AU4527599A (en) 2000-01-24

Similar Documents

Publication Publication Date Title
DE69738535T2 (de) Antivirale mittel aus pflanzenextrakten und deren verwendung zur behandlung viraler infektionen
US6039949A (en) Method of preparation and composition of a water soluble extract of the plant species uncaria
US7045159B1 (en) Antiviral substances from plant cuticular and epiciticular material
IL167391A (en) A preparation for the treatment of AIDS and related conditions
JP4558200B2 (ja) 内部寄生虫および原性動物を処置するためのオレガノ
EP0750908A1 (fr) Remede contre les maladies infectieuses
Ruffa et al. Inhibitory effect of medicinal herbs against RNA and DNA viruses
WO1997004791A1 (fr) Agent antiviral et procede de preparation
WO1994018993A1 (fr) Composition therapeutique a base d'herbes
US5980903A (en) Composition for the treatment of viral infections including HIV
EP1414475B1 (fr) Composition vegetale pour le traitement des difficultes respiratoires bronchiques
WO2000001398A1 (fr) Extrait de plante antiviral
WO2004112692A2 (fr) Compositions therapeutiques a base de plantes medicinales
AU760967B2 (en) Herbal composition for the prophylaxis and treatment of AIDS
EP0539516B1 (fr) Composition d'asphodelus permettant d'augmenter la numeration de globules blancs dans le sang
US7160561B2 (en) Herbal composition and method of treating HIV infection
JPH06211713A (ja) 抗アレルギー剤
JPH09143085A (ja) 甘草成分含有強肝剤
JPH0987195A (ja) 生薬を含有する抗エイズウイルス剤
US7282227B2 (en) Method for treating hepatitis C with evaporate of Ecballium elaterium extract
RU2804478C2 (ru) Комбинация родиолы и астрагала для лечения нейродегенеративных заболеваний
RU2187326C1 (ru) Композиция "виларин", обладающая адаптогенным и общеукрепляющим действием
KR0149986B1 (ko) 필란두스속 식물의 추출물을 포함하는 비형 간염 치료제
EA009393B1 (ru) Фармацевтическая композиция, пригодная для использования в качестве антивирусного, антибактериального средства и для стимулирования иммунной защиты
US20040234546A1 (en) Treatments using an herbal composition

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AT AU BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE HR HU ID IL IN IS JP KE KP KR LC LK LR LT LU LV MG MK MN MW MX NO NZ PL PT RO RU SE SG SI SK TR TT UA US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase