Biflavanoids and derivatives thereof as antiviral agents

Substantially purified antiviral biflavanoids robustaflavone, hinokiflavone, amentoflavone, agathisflavone, volkensiflavone, morelloflavone, rhusflavanone, succedaneaflavanone, GB-1a, and GB-2a are provided. Antiviral biflavanoid derivatives and salt forms thereof, e.g., robustaflavone tetrasulfate potassium salt, and methods for preparing the same are also disclosed. Pharmaceutical compositions which include the antiviral biflavanoids, derivatives or salts thereof are also provided alone or in combination with at least one antiviral agent such as 3TC. Also disclosed is an improved method for obtaining substantially pure robustaflavone from plant material. The biflavanoid compounds, derivatives or salts thereof of the invention may be used in a method for treating and/or preventing viral infections caused by viral agents such as influenza, e.g., influenza A and B; hepatitis, e.g., hepatitis B; human immunodeficiency virus, e.g., HIV-1; Herpes viruses (HSV-1 and HSV-2); Varicella Zoster virus (VZV); and measles. For instance, semi-synthetic hexa-O-acetate and hexa-O-methyl ether derivatives of robustaflavone have been found to be effective in a method for treating or preventing hepatitis B viral infections. Compositions which include these robustaflavone derivatives along with methods for preparing and using the same are also provided. These compositions may be used alone or in combination with at least one antiviral agent such as 3TC.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE

[0001] This application is a continuation-in-part of U.S. Ser. No. 08/842,625, filed Apr. 15, 1997 which is continuation-in-part of U.S. Ser. No. 08/668,284, filed Jun. 21, 1996, which in turn is a continuation-in-part of provisional application No. 60/000465, filed Jun. 23, 1995.

FIELD OF THE INVENTION

[0003] The present invention relates to substantially pure antiviral biflavanoids, e.g., robustaflavone, biflavanoid derivatives and salts thereof such as esters, ethers, amines, sulfates, ethylene oxide adducts, and acid salts, and pharmaceutical compositions containing the same. Representative examples include hexa-O-acetate and hexa-O-methyl ether derivatives of robustaflavone and robustaflavone tetrasodium salt. The present invention also relates to methods for extracting substantially pure robustaflavone from plant material. The present invention also relates to a method for preventing and/or treating viral infections such as hepatitis B, influenza A and B, and HIV which employ robustaflavone or derivatives thereof alone or in combination with at least one antiviral agent such as 3TC.

BACKGROUND OF THE INVENTION

[0004] Viruses, an important etiologic agent in infectious disease in humans and other mammals, are a diverse group of infectious agents that differ greatly in size, shape, chemical composition, host range, and effects on hosts. After several decades of study, only a limited number of antiviral agents are available for the treatment and/or prevention of diseases caused by viruses such as hepatitis B, influenza A and B and HIV. Because of their toxic effects on a host, many antiviral agents are limited to topical applications. Accordingly, there is a need for safe and effective antiviral agents with a wide-spectrum of anti-viral activity with reduced toxicity to the host.

[0005] Since the identification of the human immunodeficiency virus (HIV) as the causative agent of AIDS,36,46 the search for safe and effective treatments for HIV infection has become a major focus for drug discovery groups around the world. Investigations into the molecular processes of HIV have identified a number of macromolecular targets for drug design, such as HIV-1 reverse transcriptase (HIV-RT), protease and integrase enzymes, and regulatory proteins (e.g., TAT and REV). Other targets are enzymes which aid in virus attachment and fusion. HIV-RT is an essential enzyme in the life cycle of HIV, which catalyzes the transcription of HIV-encoded single-stranded RNA into double-stranded DNA. Furthermore, the RNA-dependent DNA polymerase function of HIV-RT does not have an analogous process in mammalian metabolism, and thus is a suitable target for a chemotherapeutic agent.

[0006] The hepatitis B virus (HBV) infects people of all ages. It is one of the fastest-spreading sexually transmitted diseases, and also can be transmitted by sharing needles or by behavior in which a person's mucus membranes are exposed to an infected person's blood, semen, vaginal secretions, or saliva. While the initial sickness is rarely fatal, ten percent of the people who contract hepatitis are infected for life and run a high risk of developing serious, long-term liver diseases, such as cirrhosis of the liver and liver cancer, which can cause serious complications or death.1 The World Health Organization lists HBV as the ninth leading cause of death. It is estimated that about 300 million persons are chronically infected with HBV worldwide, with over 1 million of those in the United States. The Center for Disease Control estimates that over 300,000 new cases of acute HBV infection occurs in the United States each year, resulting in 4,000 deaths due to cirrhosis and 1,000 due to hepatocellular carcinoma.2 The highest rates of HBV infections occur in Southeast Asia, South Pacific Islands, Sub-Saharan Africa, Alaska, Amazon, Bahai, Haiti, and the Dominican Republic, where approximately 20% of the population is chronically infected.3

[0007] Hepatitis B virus (HBV) infection is currently the most important chronic virus infection, but no safe and effective therapy is available at present. The major therapeutic option for carriers of HBV is alpha interferon, which can control active virus replication. However, even in the most successful studies, the response rate in carefully selected patient groups has rarely exceeded 40%.5,6 One of the reasons cited for interferon failure is the persistence of viral supercoiled DNA in the liver.7

[0008] Recently, lamivudine (3TC) has provided encouraging results against both HBV and HIV in human clinical trials. Lamivudine is approved for treatment of HIV infection, and is currently being evaluated as a treatment for HBV. A recent study of 40 HIV-HBV-coinfected patients showed dramatic decreases in the level of HBV replication following treatment with 3TC over a 12 month period, with no observable adverse effects (84). Another agent, famciclovir, an orally active derivative of the acyclic guanine derivative penciclovir (85), is approved for treatment of herpes zoster and acute recurrent genital herpes, and has also shown promising results against HBV infection in clinical trials, again with little observable toxicity (86). It is hoped that these agents will eventually provide promising treatment options for HBV infection.

[0009] Unfortunately, monotherapy of viral infections often results in selection for mutant viral strains having resistance to the antiviral drug being used. Indeed, clinical isolates of mutant HBV strains have been identified having resistance to 3TC following treatment with that agent (87,88). It is plausible that combination therapy of HBV would provide an enhanced antiviral response, while reducing the danger of resistance selection. Combination of agents has been shown to be superior in treatment of HIV relative to monotherapy (89). Thus, the identification of compounds which inhibit HBV, particularly compounds having structures other than that of the nuceloside analogues, is of critical importance in the search for effective anti-HBV regimens.

[0010] Influenza is a viral infection marked by fever, chills, and a generalized feeling of weakness and pain in the muscle, together with varying signs of soreness in the respiratory tract, head, and abdomen. Influenza is caused by several types of myxoviruses, categorized as groups A, B, and C4. These influenza viruses generally lead to similar symptoms but are completely unrelated antigenically, so that infection with one type confers no immunity against the other. Influenza tends to occur in wavelike epidemics throughout the world; influenza A tends to appear in cycles of two to three years and influenza B in cycles of four to five years. Influenza is one of the few common infectious diseases that are poorly controlled by modem medicine. Its annual epidemics are occasionally punctuated by devastating pandemics. For example, the influenza pandemic of 1918, which killed over 20 million people and affected perhaps 100 times that number, was the most lethal plague ever recorded. Since that time, there have been two other pandemics of lesser severity, the so-called Asian flu of 1957 and the Hong Kong flu of 1968. All of these pandemics were characterized by the appearance of a new strain of influenza virus to which the human population had little resistance and against which previously existing influenza virus vaccines were ineffective. Moreover, between pandemics, influenza virus undergoes a gradual antigenic variation that degrades the level of immunological resistance against renewed infection.4

[0011] Anti-influenza vaccines, containing killed strains of types A and B virus currently in circulation, are available, but have only a 60 to 70% success rate in preventing infection. The standard influenza vaccine has to be redesigned each year to counter new variants of the virus. In addition, any immunity provided is short-lived. The only drugs currently effective in the prevention and treatment of influenza are amantadine hydrochloride and rimantadine hydrochloride.11-13 While the clinical use of amantadine has been limited by the excess rate of CNS side effects, rimantadine is more active against influenza A both in animals and human beings, with fewer side effects.14,15 It is the drug of choice for the chemoprophylaxis of influenza A.13,16,17 However, the clinical usefulness of both drugs is limited by their effectiveness against only influenza A viruses, by the uncertain therapeutic efficacy in severe influenza, and by the recent findings of recovery of drug-resistant strains in some treated patients.18-22 Ribavirin has been reported to be therapeutically active, but it remains in the investigational stage of development.23,24

[0012] In influenza, amantadine and rimantadine have been shown to be moderately effective against only influenza A viruses; with amantadine having excessive side effects. Recently, strains of influenza A resistant to amantadine and rimantadine have been isolated. Accordingly, there is a need for new types of therapeutic antiviral agents against both influenza A and influenza B, as well as against HBV and HIV.

SUMMARY OF THE INVENTION

[0013] The present invention relates to substantially purified antiviral biflavanoids, derivatives and salts thereof and pharmaceutical compositions containing the same; improved methods for extracting substantially pure robustaflavone from plant material; methods for preparing derivatives and salts from antiviral biflavanoids; and methods for treating and/or preventing viral infections using the antiviral biflavanoids, derivatives and salts thereof.

[0014] The present invention provides substantially purified biflavanoids comprising robustaflavone, hinokiflavone, amentoflavone, agathisflavone, morelloflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, GB-1a, and GB-2a and pharmaceutical compositions containing the same. Scheme I illustrates the chemical structures of these biflavanoids. The biflavanoids of the invention, extractable from plant materials derived from a variety of natural sources such as Rhus succedanea and Garcinia multiflora, were found to be effective in inhibiting viral activity and may be used in a method for treating and/or preventing a broad range of viral infections such as Influenza A and B, hepatitis B and HIV-1, HSV-1, HSV-2, VZV, and measles. It has been discovered that robustaflavone effectively inhibits activity of influenza A and B viruses, hepatitis B, HIV-1, HSV-1 and HSV-2. Hinokiflavone and morelloflavone exhibited similar activity against various strains of HIV-1.

[0015] Anti-viral biflavanoid derivatives and salts and pharmaceutical compositions containing the same are also contemplated by the invention. Representative derivatives include ethers, e.g., methyl ethers, esters, amines, ethylene oxide adducts, and polymers such as trimers and tetramers of apigenin. Representative salts include sulfates and acid salts. Methods for preparing these derivatives and salts are also provided. It has been discovered for instance that salts of robustaflavone, e.g., robustaflavone tetrasulfate potassium salt and robustaflavone tetrasodium salt, effectively inhibit hepatitis B activity. Furthermore, robustaflavone hexa-O-acetate and hexa-O-methyl ether derivatives of robustaflavone were found to be not only potent inhibitors of HBV replication but with greatly reduced cytotoxicity. Scheme I illustrates several examples of biflavanoid derivatives.

[0016] Improved methods for extracting robustaflavone from plant material are also provided. According to one method, a substantially pure robustaflavone in greater yields can be obtained through the use of a particular solvent mixture comprising toluene/ethanol/pyridine. The improved extraction method eliminates the use of benzene and requires smaller volumes of pyridine from the prior reported methods.

[0017] A second improved method for purification of robustaflavone is also provided which involves acetylation of the extracted pigment to produce acetates of the pigments. Robustaflavone acetate is then purified by recrystallization and converted to robustaflavone by hydrolysis. This method eliminates the use of pyridine in column chromatography and is ideal for large scale extraction of robustaflavone.

[0018] Finally, a method for treating and/or preventing viral infections using antiviral biflavanoids alone or in combination with one or more other antiviral agents is described. Representative viral infections include influenza A and B viruses, hepatitis B and human immunodeficiency virus (HIV-1), HSV-1, HSV-2, VZV, and measles.

[0019] Accordingly, it is an object of the invention to provide substantially purified antiviral biflavanoids robustaflavone, hinokiflavone, amentoflavone, agathisflavone, morelloflavone, rhusflavanone, succedaneaflavanone, GB-1a, and GB-2a.

[0020] It is another object of the invention to provide antiviral derivatives and salt forms of biflavanoids robustaflavone, hinokiflavone, amentoflavone, agathisflavone, morelloflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, GB-1a, and GB-2a as well as method of preparation thereof. A representative example of an antiviral biflavanoid derivatives include robustaflavone tetrasulfate potassium salt, robustaflavone tetrasodium salt, robustaflavone hexa-O-acetate, and robustaflavone hexa-O-methyl ether.

[0021] It is yet another object of the invention to provide pharmaceutical compositions which include at least one antiviral biflavanoids such as robustaflavone, hinokiflavone, amentoflavone, agathisflavone, morelloflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, GB-1a, GB-2a, derivatives or salts thereof. Pharmaceutical compositions including an effective amount of at least one antiviral biflavanoid in combination with at least one other antiviral agent, e.g. robustaflavone with penciclovar or lamivudine (3TC), for use in combination antiviral therapy are also contemplated.

[0022] It is a further object of the invention to provide an improved method for obtaining substantially pure robustaflavone and in greater yields than prior procedures.

[0023] It is yet a further object of the invention to provide a method for treating and/or preventing viral infections which comprises administering an antiviral effective amount of a biflavanoid. Representative viral infections are caused by viral agents such as influenza, e.g., influenza A and B; hepatitis, e.g., hepatitis B; human immunodeficiency virus, e.g., HIV-1; HSV-1, HSV-2, VZV, and measles.

[0024] These and other objects of the invention will become apparent in light of the detailed description below. 1

DESCRIPTION OF THE DRAWINGS

[0025] FIG. 1 illustrates the effect of treatment with robustaflavone in DMSO on mean arterial oxygen saturation (mean SaO2 (%)) in Influenza A virus-infected mice as described in Example 11.

[0026] FIG. 2 illustrates the effect of treatment with robustaflavone in DMSO on mean lung scores in Influenza A virus-infected mice as described in Example 11.

[0027] FIG. 3 illustrates the effect of treatment with robustaflavone in DMSO on mean lung weights in Influenza A virus-infected mice as described in Example 11.

[0028] FIG. 4 illustrates the effect of treatment with robustaflavone in DMSO on mean virus titers in Influenza A virus-infected mice as described in Example 11.

[0029] FIG. 5 illustrates the effect of treatment with robustaflavone in CMC on mean arterial oxygen saturation (mean SaO2 (%)) in Influenza A virus-infected mice as described in Example 11.

[0030] FIG. 6 illustrates the effect of treatment with robustaflavone in CMC on mean lung scores in Influenza A virus-infected mice as described in Example 11.

[0031] FIG. 7 illustrates the effect of treatment with robustaflavone in CMC on mean lung weights in Influenza A virus-infected mice as described in Example 11.

[0032] FIG. 8 illustrates the effect of treatment with robustaflavone in CMC on mean virus titers in Influenza A virus-infected mice as described in Example 11.

DETAILED DESCRIPTION OF THE INVENTION

[0033] All references and patents cited herein are hereby incorporated by reference in their entirety.

[0034] In one embodiment of the invention, substantially pure biflavanoids robustaflavone, hinokiflavone, amentoflavone, agathisflavone, morelloflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, GB-1a, and GB-2a, derivatives and salts of the biflavanoids, and pharmaceutical compositions containing the same are disclosed. Methods for extracting and isolating the biflavanoids were previously reported.28,37,39,40,53-55 Moreover, methods for preparing derivatives such as the acetate37,38 and methyl ethers39,40 for several of these biflavanoids are also reported. Representative methods for preparing biflavanoid derivatives are illustrated in the examples below. Applicants have determined that these biflavanoids, especially robustaflavone, were surprisingly effective in inhibiting one or more activities of viruses such as Influenza A and B, hepatitis B and HIV-1, HSV-1, HSV-2, VZV, and measles.

[0035] Approximately 100 biflavanoids have been isolated to date, since the first biflavanoid, a biflavone, was isolated in 1929 by Furukawa from ginkgo biloba L. as a yellow pigment.44,45,61 Biological activities of several biflavanoids, such as ginkgetin, have been reported. For instance, peripheral vasodilatation, anti-bradykinin, and anti-spasmogenic activities have been observed.48,62 Garcinikolin stimulates RNA synthesis in rat hepatocyte suspensions.57 Also, agathisflavone, kolaviron, GB-1 and GB-2 have hepatoprotective activity.33,49 Hinokiflavone, kayaflavone, bilobetin, lophirone A, lophiraic acid, and sotetusflavone demonstrate inhibitory action on the genome expression of the Epstein-Barr virus (EBV).51,52,60 GB-1 exhibits molluscicidal activity,65 while daphnodorin A, daphnodorin B, and daphnodorin D possess antimicrobial activity.34 Hinokiflavone exhibits cytotoxicity against tissue cultured cells of human mouth epidermoid carcinoma (KB).56 Amentoflavone and morelloflavone exhibit an inhibitory effect on lipid peroxidation,41,59,66 and kolaviron produced hypoglycemic effects.50 None of these references, however, disclose or suggest that robustaflavone, hinokiflavone, morelloflavone, amentoflavone, agathisflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, GB-1a and GB-2a, especially robustaflavone and its tetrasulfate potassium salt, have an inhibitory effect against at least one of influenza, e.g., influenza A and B; hepatitis, e.g., hepatitis B; human immunodeficiency virus, e.g., HIV-1; HSV-1, HSV-2, VZV, and measles.

[0036] In another embodiment of the invention, an improved method for extracting substantially pure robustaflavone from natural sources is also provided. Robustaflavone, 1, a naturally occurring biflavanoid, was previously isolated, purified, and identified from the seed-kemels of Rhus succedanea.25 Other sources of robustaflavone include: seed kernel of Rhus succedanea L.;25 leaves of Selaginella lepidophylla;27 leaves of Anacardium occidentale;28 leaves and branches of Podocarpus neriifolius D. Doa;29 Selaginella denticulata;30 and Selaginella willdenowii.31

[0037] The drupes of wax-tree, Rhus succedanea L (Anacardiaceae), are of great economic importance in that they yield Japan wax. Earlier work on this species has shown the presence of fustin and fisetin in the wood, rhoifolin in leaves, japanic acid in the wax, and ellagic acid, fatty acids, and flavanoids in the seed kernels. Further studies of the pigment in the seed kernels of wax-tree led to the isolation of eight biflavanoids, four of which were new. Concentration of the ethanol extract of the seed kernels yielded, successively, fractions of ellagic acid, pigment A (hinokiflavone and robustaflavone) and pigment B (amentoflavone). Further concentrations gave a crude yellow pigment C which, when subjected to silica gel column chromatography, afforded fractions CI (rhusflavanone, succedaneaflavanone and neorhusfiavanone), CII (rhusflavone), and CIII (agathisflavone).

[0038] A prior method for extracting and isolating substantially pure robustaflavone from plant material was reported.55 This method, however, used large quantities of benzene and pyridine which is undesirable for use in large scale extractions and produced mediocre yields of robustaflavone. The Applicants discovered improved extraction methods which eliminates benzene, greatly reduces or eliminates the amount of pyridine used and produces at least double the quantities of substantially pure robustaflavone compared to the prior method. In one method of the invention, solvent mixture comprising toluene/ethanol/formic acid at a volume ratio ranging about 10-30:2-10:1, preferably about 20:5:1, was found to be useful as a developing solvent mixture for the dry column procedure. This particular solvent mixture was found to be especially useful in large scale extractions.

[0039] In a second method for purification of robustaflavone, extracted pigment is acetylated followed by purification and hydrolysis of the robustaflavone acetate to produce pure robustaflavone. This method eliminates the use of pyridine in column chromatography and is ideal for large scale isolation of purified robustaflavone. According to the second method, pigment A was converted to the acetate with an acylating agent in solution. Suitable, but non-limiting, acylating agents include acetic anhydride and acetyl chloride. The preferred acylating agent is acetic anhydride. In general, excess amounts of acylating agent relative to pigment A are used. Suitable, but non-limiting, solvents for use in this second method include any suitable solvent generally used for acetylation reactions including pyridine and triethylamine. In practicing this invention, pyridine is the preferred solvent.

[0040] The acetylation reaction is generally performed at room temperature until completion is reached as ascertained by the usual organic chemical methods such as thin layer chromatography or Gas liquid chromatography. Thereafter, the crude acetate is precipitated out in water and collected by filtration. The crude precipitate is then washed with water and recrystallized out from a suitable solvent or solvent mixture. In practicing this invention, robustaflavone acetate is preferrably recrystallized from a 9:1 solvent mixture of methylene chloride and ethyl acetate. If desired, additional acetate may be recovered from the mother liquor by column chromatography. The purified robustaflavone acetate is then hydrolyzed with excess aqueous alkaline solution such as 2M aqueous sodium hydroxide solution. In practicing this invention, robustaflavone acetate hydrolysis mixture is preferably to a temperature ranging between about 30 to about 100° C., preferably around 60° C., for a time sufficient to hydrolyze off the acetate groups. The alkaline reaction mixture is then cooled in an ice bath and acidified to about pH 3.0 with excess amounts of aqueous acid solution, e.g., 3N hydrochloric acid, to precipitate out robustaflavone as a yellow solid.

[0041] Examples of extraction via the improved extraction methods of the invention are illustrated in the examples below.

[0042] In yet another embodiment of the invention, a method is provided for treating and/or preventing viral infections in mammals comprising administering an antivirally effective amount of a biflavanoid such robustaflavone, hinokiflavone, amentoflavone, agathisflavone, morelloflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, GB-1a, and GB-2a. In practicing this invention, administration of robustaflavone or derivatives thereof is preferred. Examples of mammals include humans, primates, bovines, ovines, porcines, felines, canines, etc. Examples of viruses may include, but not be limited to, HIV-1, HIV-2, herpes simplex virus (type 1 and 2) (HSV-1 and 2), varicella zoster virus (VZV), cytomegalovirus (CMV), papilloma virus, HTLV-1, HTLV-2, feline leukemia virus (FLV), avian sarcoma viruses such as rous sarcoma virus (RSV), hepatitis types A-E, equine infections, influenza virus, arboviruses, measles, mumps and rubella viruses. More preferably the compounds of the present invention will be used to treat a human infected with hepatitis and/or influenza virus. Preferably the compounds of the present invention will also be used to treat a human exposed or infected (i.e., in need of such treatment) with the human irnmunodeficiency virus, either prophylactically or therapeutically.

[0043] Antiviral biflavanoids and derivatives thereof may be formulated as a solution of lyophilized powders for parenteral administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use. The liquid formulation is generally a buffered, isotonic, aqueous solution. Examples of suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or in buffered sodium or ammonium acetate solution. Such formulation is especially suitable for parenteral administration, but may also be used for oral administration. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium choride or sodium citrate.

[0044] Alternatively, the compounds of the present invention may be encapsulated, tableted or prepared in an emulsion (oil-in-water or water-in-oil) syrup for oral administration. Pharmaceutically acceptable solids or liquid carriers, which are generally known in the pharmaceutical formulary arts, may be added to enhance or stabilize the composition, or to facilitate preparation of the composition. Solid carriers include starch (corn or potato), lactose, calcium sulfate dihydrate, terra alba, croscarmellose sodium, magnesium stearate or stearic acid. talc, pectin, acacia, agar, gelatin, maltodextrins and microcrystalline cellulose, or collodial silicon dioxide. Liquid carriers include syrup, peanut oil, olive oil, corn oil, sesame oil, saline and water. The carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax. The amount of solid carrier varies but, preferably, will be between about 10 mg to about 1 g per dosage unit.

[0045] The dosage ranges for administration of biflavanoids or derivatives thereof are those which produce the desired affect whereby symptoms of infection are ameliorated. For example, as used herein, a pharmaceutically effective amount for influenza or hepatitis infection refers to the amount administered so as to maintain an amount which suppresses or inhibits circulating virus throughout the period during which infection is evidenced such as by presence of anti-viral antibodies, presence of culturable virus and presence of viral antigen in patient sera. The presence of anti-viral antibodies can be determined through use of standard ELISA or Western blot assays for example. The dosage will generally vary with age, extent of the infection, the body weight and counterindications, if any, for example, immune tolerance. The dosage will also be determined by the existence of any adverse side effects that may accompany the compounds. It is always desirable, whenever possible, to keep adverse side effects to a minimum.

[0046] One skilled in the art can easily determine the appropriate dosage, schedule, and method of administration for the exact formulation of the composition being used in order to achieve the desired effective concentration in the individual patient. However, the dosage can vary from between about 0.001 mg/kg/day to about 150 mg/kg/day, but preferably between about 1 to about 50 mg/kg/day.

[0047] The pharmaceutical composition may contain other pharmaceuticals in conjunction with biflavanoids and derivatives thereof to treat (therapeutically or prophylactically) antiviral infections. For example, other pharmaceuticals may include, but are not limited to, other antiviral compounds (e.g., AZT, ddC, ddI, D4T, 3TC, acyclovir, gancyclovir, fluorinated nucleosides and nonnucleoside analog compounds such as TIBO derivatives, nevirapine, saquinavir, a-interfon and recombinant CD4), immunostimulants (e.g., various interleukins and cytokines), immunomodulators and antibiotics (e.g., antibacterial, antifungal, anti-pneumocysitis agents).

[0048] The following examples are illustrative and do not serve to limit the scope of the invention as claimed. In these examples, eleven biflavanoids, amentoflavone (1), agathisflavone (2), robustaflavone (3), hinokiflavone (4), volkensiflavone (5), morelloflavone (7), rhusflavanone (9), succedaneaflavanone (11), GB-1a (13), GB-1a 7″-O-b-glucoside (15), and GB-2a (16), isolated from Rhus succedanea and Garcinia multiflora, and their methyl ethers, acetate and sulfate potassium salt, volkensiflavone hexamethyl ether (6), morelloflavone heptamethyl ether (8), rhusflavanone hexaacetate (10) succedaneaflavanone hexaacetate (12), GB-1a hexamethyl ether (14) and robustaflavone tetrasulfate potassium salt were evaluated for their antiviral activities. The inhibitory activities against HIV-1 RT and various viruses including herpes viruses (HSV-1, HSV-2, HCMV, and VZV), and respiratory viruses (influenza A, influenza B, RSV, parainfluenza 3, adenovirus 5, and measles) were investigated.

EXAMPLE 1 Extraction and Isolation of Biflavanoids Isolation of Compounds

[0049] Compounds tested were isolated from the seed kernels of Rhus succedanea obtained from Fukkuoka, Japan, and also from the heartwood of Garcinia multiflora collected in Taiwan.

[0050] Amentoflavone (1),53 agathisflavone (2),54 robustaflavone (3),55 hinokiflavone (4),55 rhusflavanone (9),37 and succedaneaflavanone (11)28 were isolated from Rhus succedanea. Rhusflavanone hexaacetate (10) and succedaneaflavanone hexaacetate (12) were prepared directly from compounds (9) and (11), respectively.37,38 Volkensiflavone (5), morelloflavone (7), GB-1a (13), GB-1a glucoside (15), and GB-2a (16) were isolated from Garcinia multiflora.39,40 Volkensiflavone hexamethylether (6), morelloflavone hexamethylether (8), and GB-1a hexamethylether (14) were prepared from compounds (5), (7), and (13), respectively.39,40 Robustaflavone tetrasulfate potassium salt (17) was prepared from robustaflavone (3).

[0051] In this example, two procedures for isolating robustaflavone are described. In the first procedure, robustaflavone was isolated by a dry-column method using benzene/pyridine/formic acid (20:5:1) as developing solvent, following an earlier reported procedure.25 In order to eliminate the use of benzene and large quantities of pyridine, an improved procedure was developed wherein benzene and pyridine are replaced with other solvents. The solvent mixture of toluene/ethanol/formic acid in the ratio of 20:5:1 was used as the developing solvent in the dry-column procedure. Hinokiflavone was eluted completely from the dry-column and robustaflavone retained in the column. A mixture of ethanol and pyridine in the ratio of 4:1 was then used to elute robustaflavone from the column.

[0052] Extraction of Biflavanoids from Rhus succedanea. The seeds (16 kg) of Rhus succedanea obtained from Fukuoka, Japan were coarsely powdered and defatted with benzene. The defatted seeds were exhaustively extracted with boiling 95% EtOH (150 L). The combined EtOH extracts were concentrated in vacuo. The yellow pigments obtained during the concentration were filtered to yield crude pigment A (yield 0.2%) and pigment B (yield 0.2%). successively. Further concentration yielded yellow pigment C (ca. 2%).

[0053] Isolation of Robustaflavone from Pigment A. One gram of pigment A dissolved in 10 mL of pyridine was mixed with 5 g of silica gel (Kiselgel nach Stahl Type 60 Merck) and evaporated in vacuo to remove pyridine. The dried yellow powder obtained was packed on the top of a silica gel column (SiO2100 g, 4×20 cm). The solvent mixture (400 mL) of benzene/pyridine/formic acid (40:10:2) was passed through the column. The column was sliced into seven bands (bands 1˜7 from top to bottom). Extraction of the yellow band 4 with EtOAc and subsequent concentration of the extract yielded yellow crystals (200 mg), robustaflavone, which were recrystallized from pyridine-water, m.p. 350-352° C. (dec.). Mg-HCl test (orange red color), FeCl3/EtOH test (brown color). IR cm−1 (KBr): 3300 (OH), 1655, 1645 (CO), 1610, 1570, 1510, 1505, 1485 (aromatic ring), UVlmax (MeOH) nm (log &egr;): 255 (4.71), 275 (4.44), 300 (4.42), 347 (4.49), lmax (NaOAc-MeOH) nm (log &egr;): 257 (4.66), 277 (4.48), 313 (sh, 4.41), 378 (4.38), lmax (AlCl3-MeOH) nm (log &egr;): 254 (4.80), 278 (4.45), 300 (4.45), 352 (4.50), 388 (4.43); NMR (DMSO-d6) (60 MHz) dppm: 7.87 (1H, d, J=2 Hz, H-2′), 7.94 (1H, dd, J=2 Hz, 9 Hz, H-6′), 7.09 (1H, d, J=9 Hz, H-5′), 7.97 (2H, d, J=9 Hz, H-2″′, 6″′), 7.03 (2H, d. J=9 Hz, H-3″′, 5″′), 6.23 (1H, d, J=2 Hz, H-6), 6.52 (1H, d, J=2 Hz, H-8), 6.68 (1H, s, H=8″), 6.80 (1H, s, H-3 or H3″), 6.83 (1H, s, H-3, or 3″), 13.53 (1H, s, HO-5), 13.28 (1H, s, HO-5″), 11.23˜8.63 (4H, br., 4×OH), Anal, Calcd. for C30H18O10.H2O: C, 64.75; H, 3.62, Found: C, 64.5 1; H, 3.83.

Improved Procedure for Isolating Robustaflavone (Method No. 1)

[0054] Pigment A (10 g) was dissolved in 50 mL of pyridine. The solution was added to 25 g of silica gel and thoroughly mixed. The pyridine was removed under reduced pressure using a rotary evaporator and the dry mixture ground to a fine particle size. To a 600 mL fritted filter funnel, incorporating a coarse porosity sinter with a disc of filter paper placed over the sinter, was added 250 g of silica gel. The adsorbed Pigment A was then carefully placed and spread on the top of the silica gel in the funnel. The solvent system of toluene/ethanol/formic acid (40:10:2) (2.5 L) was passed through the funnel to remove the hinokiflavone. The eluent was collected and concentrated to provide 2.01 g of a yellow solid which was identified as hinokiflavone and a trace of robustaflavone.

[0055] The silica gel in the fritted funnel was allowed to dry out overnight. The top layer containing the adsorbed Pigment A was then scraped off the remaining silica gel and placed into a fritted filter fuinel of coarse porosity containing a disc of filter paper. The silica gel containing the adsorbed pigment A was then eluted using a mixture of toluene/ethanol/formic acid (40:10:2) (2.5 L), and then ethanol/pyridine (4:1) (4.5 L). The first eluting solution was concentrated to afford 1.1 g of a yellow solid which was identified as a mixture of robustaflavone and hinokiflavone, the major component being robustaflavone. The second eluting solution (ethanol/pyridine 4:1) was concentrated to afford robustaflavone (5.65 g). TLC, NMR, MS, and elemental analysis support these findings. NMR (H-NMR, 13C-NMR, COSY and HETCOR NMR: see Table 1).

Improved Procedure for Isolating Robustaflavone (Method No. 2)

[0056] Pigment A, a mixture of robustaflavone and hinokiflavone, was converted to the hexaacetate and pentacetate, respectively, with acetic anhydride in pyridine. The resulting robustaflavone hexaacetate was purified by recrystallization from the solvent mixture of dichloromethane and ethyl acetate to obtain robustaflavone hexaacetate in the yield of 55%. The mother liquor could be further purified by column chromatography to offer additional amounts of robustaflavone hexacetate.

[0057] Robustaflavone hexaacetate was hydrolyzed with 2M aqueous sodium hydroxide at 60° C. for 30 min. The resulting yellow solution was further stirred at room temperature for additional 2.5 and then acidified with hydrochloric acid (˜3N) to pH 3 at 0° C. to precipitate robustaflavone as a yellow solid in a yield of 89.6%.

Robustaflavone Hexaacetate

[0058] To Pigment A (8.84 g) were added 50 mL of pyridine and 50 mL acetic anhydride. The resulting solution was allowed to stand at room temperature for 2 days. The reaction mixture was then poured into ice water (800 mL) with stirring. The precipitate was collected by filtration and rinsed with ice water to remove pyridine and acetic anhydride to afford an ivory solid, 12 g (yield 92.5%), which was recrystallized from the solvent mixture of dichloromethane and ethyl acetate in the ratio of 9:1 to yield robustaflavone hexaacetate as a white powder, 6.32 g (55% yield), m.p. 197-198° C., APCI-MS m/z 791.1 [M+H]; H-NMR &dgr;CDC13 7.93 (d, J=8.6 Hz, 1H, H-6″′), 7.92 (dd, J-8.7 HZ, 2.4 HZ, 2H, H-2′,6′), 7.85 (d,=2.1 HZ, 1H, H-2″′), 7.43 (d, J=2.4 Hz, 1H, H-5″′), 7.31 (d, J=2.1 Hz, 1H, H=6″), 7.28 (d, J=2.1 Hz, 1H, H-8″), 6.86 (d, J=2.1 Hz, 1H, H-6″), 6.68 & 6.66 (each s, 1H, H-3,3″), 2.44, 2.36, 2.34, 2.21, 2.13 & 2.06 (each, s, 3H, 6x OAc); HPLC Rt 13.31 (column: Zorbax 4.6 mm×250 mm, mobile phase: hexane/ethyl acetate 1.2).

Robustaflavone—Hydrolysis of Robustaflavone Hexaacetate

[0059] To robustaflavone hexaacetate (1 g) was added 10 mL of 2M aqueous NaOH and the mixture was heated to 60° C. for 30 min. The resulting yellow solution was further stirred at room temperature for an additional 2.5 and then acidified with 3 N of hydrochloric acid to pH 3 at 0° C. to precipitate robustaflavone. The yellow precipitate was collected by filtration and washed with ice water to obtain robustaflavone as yellow solid, 610 mg (yield 89.6%), m.p. 197-198° C.; APCI-MS m/z 539.3 [M+H]+; H—NMR and 13C—NMR identical to the previous data; HPLC Rt 42.2 (column: C18, 4.6 mm×15 cm, mobile phase: 20% MeOH/80% of 1% TFA in water to 100% MeOH, gradient).

Characterization of Robustaflavone

[0060] Robustaflavone was recrystallized from pyridine/water, mp. 350-352° C. (dec.). The compound gave an orange-red color in the Mg-HCl test and a brown color with alcoholic FeCl3. The IR spectrum showed a broad hydroxyl absorption at 3250 cm−1 and a conjugated carbonyl absorption at 1650 cm−1. The UV spectrum in MeOH exhibited four maxima in the region of 347 (log &egr;4.38), 300 (4.42), 275 (4.44) and 255 (4.71) nm, and underwent a bathochromic shift on addition of NaOAc or AlC3. The UV spectrum in AlCl3-MEOH was similar to that of in AlCl3-MEOH upon addition of HCl, indicating the presence of OH groups at the 5,7 and 4′positions, and the absence of an o-dihydroxyl group.

[0061] The NMR spectrum (60 MHz) of robustaflavone exhibited six OH groups at &dgr;13.53 (s, 1H), 13.28 (s, 1H) and 11.23-8.63 (br, 4H); the four protons in the 1,4-disubstituted benzene ring appeared at &dgr;7.97 (d, J=9 Hz, 2H) and 7.03 (d, J=9 Hz, 2H); the three protons in the 1,3,4-trisubstituted benzene ring appeared at &dgr;7.87 (d, J=9 Hz, 1H), 7.94 (dd, J=2 Hz, 9 Hz, 1H) and 7.09 (d, J=9 Hz, 1H); two aromatic protons appeared as meta-coupled doublets (J=2 Hz) at 6.23 (1H) and 6.52 (1H); three isolated protons appeared at d 6.83(s), 6.80(s) and 6.68(s) respectively. The above evidence suggested that the structure of the compound was composed of two apigenin units joined by an interflavonyl linkage of C3″′-C6, i.e. robustaflavone, an isomer of amentoflavone. This was further supported by examination of its acetate and methyl ether. Acetylation with pyridine/Ac2O yielded robustaflavone hexaacetate (3a) as colorless needles, m.p. 199-200° C. Methylation with Me2SO4/K2CO3 in dry acetone afforded a colorless compound, robustaflavone hexamethylether (3b), m.p. 300-305° C., C36H30O10, M30 m/z 622. (The induced change in the chemical shifts (ppm) owing to the addition of Eu(fod)3 on compound (3b) is represented by an S-value.35) The S-values of MeO-II-5 and MeO-I-5 were 10.85 ppm (largest) and 2.17 ppm respectively, whereas H-I-8 was 0.34 ppm, indicating the presence of a linkage of CII-3″′-CI-6 as structure (3b) which was characterized as hexa-O-methylrobustaflavone by comparison with an authentic sample (TLC, IR, NMR and MS).35 1 TABLE I Assignment of 13C-1H HETCOR NMR 13C-&dgr;ppm H-&dgr;ppm I-2 164.11a >C═ II-2 163.86a >C═ I-3 102.86 ═CH 6.81(s) II-3 116.10 ═CH 6.84(s) I-4 181.74b >CO II-4 181.83b >CO I-5 161.20c ═C—OH 13.02(s) II-5 159.61c ═C—OH 13.23(s) I-6 108.89 ═C< II-6 98.82 ═CH 6.20(d,J=2.0 Hz) I-7 162.06d ═C—OH 10.82-10.00(br) II-7 163.65d ═C—OH 10.82-10.00(br) I-8 93.44 >CH 6.65(s,1H) II-8 94.05 >CH 6.49(d,J=2.0 Hz) I-9 161.46 ═C—O— II-9 157.5 ═C—O— I-10 103.57 ═C< II-10 103.72 ═C< I-1′ 121.22 ═C< II-1′ 120.89 ═C< I-2′ 128.55 ═CH 7.99(d,J=8.8 Hz) II-2′ 130.87 ═CH 7.79(d,J=2.2 Hz) I-3′ 116.01 ═CH 6.96(d,J=8.8 Hz) II-3′ 120.86 ═C< I-4′ 156.35 ═C—OH 10.40(br) II-4′ 159.07 ═C—OH 10.20(br) I-5′ 116.01 ═CH 6.96(d,J=8.8 Hz) II-5′ 102.86 ═CH 7.05(d,J=8.7 Hz) I-6′ 128.55 ═CH 7.99(d,J=8.88 Hz) II-6′ 127.57 ═CH 7.93(dd,J=8.7 & 2.2Hz) Assignments bearing the same alphabetical superscript in the spectrum may be reversed.

[0062] The high resolution CI mass spectrum provided an M+H ion, m/z 539.096993, C30H19O10, which requires 539.097821572. The infrared spectrum exhibited a broad hydroxyl absorption at 3250 cm−1 and a conjugated carbonyl absorption at 1650 cm−1. The UV spectrum in MeOH contained four maxima in the region of 345 (log &egr;4.49), 300 (4.42), 275 (4.44) and 255 (4.71 nm, and underwent a bathochromic shift on addition of NaOAc or AlCl3. The UV spectrum in AlCl3-MeOH was similar to that obtained in AlCl3-MeOH on addition of HCl, indicating the presence of OH groups in the 5,7 and 4′ positions, and the absence of an o-dihydroxy group.26 [lNaOAc-MeOH (log &egr;) 378 (4.38), 313 (sh 4.41), 277 (4.48), 257(4.66) nm; lAlC3-MeOH (log &egr;) 388 (4.43), 352 (4.50), 300 (4.45), 278 (4.45), 254 (4.80 nm).

[0063] The NMR (300 MHz) spectrum of robustaflavone contained six OH groups at &dgr;13.25 (1H, s), 13.02 (1H, s), 10.83 (1H, s), 10.40 (1H, s), 10.4˜10.9 (2H, br.); the four protons in the 1,4-disubstituted benzene ring at &dgr;7.98 (2H, d, J=8.88 Hz, H-2″′, 6″′and 6.96 (2H, d, J=8.88 Hz, H-3″′, 5″′); the three protons in the 1,3,4-trisubstituted benzene ring at &dgr;7.93 (1H, dd., J=8.7 Hz and 2.2 Hz, H-6′), 7.79 (1H, d. J=2.2 Hz, H-2′) and 7.05 (1H, d, J=8.7 Hz, H-5′); the five aromatic protons at &dgr;6.84 (1H, s, H-3′), 6.8 (1H, s, H-3″), 6.65 (1H, s, H-8″), 6.49 (1H, d, J=2.0 Hz, H-8) and 6.20 (1H, d, J=2 Hz, H-6).

EXAMPLE 2 General Procedure for Synthesizing O-Acyl Biflavanoids

[0064] Procedure 1: To a solution of biflavanoid in anhydrous dichloromethane containing 20% dry pyridine is added an appropriate acyl chloride or anhydride at 0° C. or at room temperature. The mixture is allowed to stand overnight, and the volatiles are evaporated in vacuo. Alternatively, the mixture is poured into water and extracted with chloroform. The organic layer is washed with water and brine, dried over anhydrous sodium sulfate, and concentrated in vacuo. The residue is chromatographed on preparative TLC or a silica gel column to afford the product.78

[0065] Procedure 2: Preparation of acetate: Biflavanoid is reacted with acetic anhydride in pyridine at room temperature overnight. The reaction mixture is poured into ice water. The precipitate is filtered and washed with cold 1% hydrochloric acid and then with water to give biflavanoid acetate.37

[0066] Rhusflavanone hexaacetate: Acetylation of rhusflavanone (200 mg) with Ac2O/Pyridine at room temperature for 20 h gave hexaacetate (110 mg) as micro needles, m.p. 130-131° C., EIMS M− m/z 794; IR cm−1 (KBr) 1770 (acetoxy CO), 1688 (flavanone CO), 1603, 1560, 1510 and 1490 (arom.); H—NMR &dgr; (CDCl3): 2.02 (3H, s, AcO-7″), 2.10 (3H, s, AcO-7), 2.15 (3H, s, AcO-5), 2.28 (3H, s, AcO-4″′), 2.32 (3H, s, AcO-4′), 2.40 (3H, s, AcO-5″), 2.85-3.06 (4H, m, H-3.3″), 5.45-5.35 (2H, m. H-2, 2″), 6.71 (1H, s, H-6″), 6.91 (1H, s, H-8), 7.14 (2H, d, J=9 Hz, H-3″′, 5″′), 7.17 (2H, d, J=9 Hz, H-3′, 5′), 7.44 (2H, d, J=9 Hz, H-2″′, 6″′), 7.55 (2H, d, J=9 Hz, H-2′, 6′).

[0067] Succedaneaflavanone hexaacetate: Acetylation of succedaneaflavanone by Procedure No. 2 produced succedaneaflavanone hexaacetate as white needles. m.p. 252-255° C. (from CHCl3-MeOH), IR cm−1 (KBr): 1770 (OAc), 1688 (flavanone CO), 1613, 1560, 1510 (arom.); H—NMR &dgr; (CDCl3): 2.10 (6H, s, AcO-7, 7″), 2.17 (6H, s, AcO-5,5″), 2.33 (6H, s, AcO-4′, 4″′), 2.83-3.27, 4H, m, H-3, 3″), 5.63 (2H, dd, J=12 Hz, 4 Hz, H-2, 2″), 6.97 (2H, s, H-8, 8″), 7.25 (4H, d, J=8 Hz, H-3′, 5′, 3″, 5″), 7.58 (4H, d, J=8 Hz, H-2′, 6′, 2″′, 6″′).

[0068] Robustaflavone hexaacetate: A solution of robustaflavone (100 mg, 0.186 mmol) in a mixture of pyridine and acetic anhydride (1 mL each) was allowed to stand at room temperature for 72 h. The solution was poured into ice water, and the resulting white precipitate was collected on a fritted glass funnel and rinsed with water (123 mg, 84.4%). Following recrystallization from EtOAc/MeOH (1:1, 1.5 mL), 88.5 mg of off-white microcrystalline material was obtained. Physical and spectral properties were identical to those previously reported in the literature.55

EXAMPLE 3 General Procedure for Synthesizing Biflavanoid Ethers

[0069] Preparation of biflavanoid alkyl ethers: To a mixture of biflavanoid and Ag2O (catalytic amount) in DMF is added a corresponding alkyl halide at 10-12° C. After stirring for 2.5-4 h, the reaction mixture is kept in a refrigerator overnight. The catalyst is filtered, and the filtrate is washed with water and brine and then concentrated in vacuo. The residue is purified by column chromatography on silica gel to yield the product.78

[0070] Preparation of biflavanoid methyl ethers: Biflavanoid is dissolved in anhydrous acetone and potassium carbonate and dimethyl sulfate are added. The solution is refluxed for 4 h. The precipitate (potassium carbonate) is filtered and the filtrate is concentrated under vacuum. The residue is dissolved in chloroform and washed with brine, dried with magnesium sulfate and concentrated under vacuum. The resulting crude product is purified by silica gel column chromatography or preparative thin layer chromatography and then recrystallized with ethyl acetate, ethanol, or chloroform to afford biflavanoid methyl ethers.37

[0071] Volkensiflavone hexamethyl ether (6)

[0072] Volkensiflavanone (200 mg) was dissolved in 30 mL of anhydrous acetone, and 4 g of potassium carbonate and 3 mL of dimethyl sulfate were added. The solution was refluxed for 4 h. The precipitate (potassium carbonate) was filtered and the filtrate was concentrated under vacuum. The reddish brown oily residue was dissolved in 15 mL of chloroform and the chloroform solution was washed with brine twice and then water. The chloroform layer was dried with magnesium sulfate and concentrated under vacuum. The residue was purified by silica gel column chromatography and eluted with the mixture of toluene and ethyl acetate in the ratio of 1:1. The eluent was concentrated under vacuum and the residue was recrystallized with methanol/chloroform to obtain 135 mg of white crystals, m.p. 258-260° C., EIMS M+ m/z 624; IR cm−1 (KBr): 2900, 2950, 2850 (OMe), 1680 (flavanone CO), 1645 (flavone CO), 1600, 1580, 1510 and 1490 (arom.) H—NMR d (CDCl3): 3.93 (3H, s, OMe), 3.87 (3H, s, OMe); 3.83 (6H, s, OMe), 3.77 (3H, s, OMe), 3.67 (3H, s, OMe), 4.90 (1H, d, J=12 Hz, H-3), 5.8 (1H, d, J=12 Hz, H-2), 6.22 (1H, d, J=2 Hz, H-6), 6.23 (1H, s, H-6″′), 6.32 (1H, d, J=2 Hz, H-8), 6.50 (1H, s, H-3″), 6.63 (1H, s, J=9 Hz, H-3′, 5′), 6.87 (2H, d, J=9 Hz, H-3″′, 5″′), 7.13 (2H, d, J=9 Hz, H-2′, 6′), 7.70 (2H, d, J=9 Hz, H-2″′, 6″).

[0073] GB-1a hexamethyl ether (14)

[0074] GB-1a (200 mg) was methylated by the method described above. The resulting crude methyl ether was purified by preparative thin layer chromatography using ethyl acetate as developing solvent. The band at Rf 0.35 was scraped off and extracted with ethyl acetate. The ethyl acetate extract was concentrated under vacuum and the residue was recrystallized from the solvent mixture of acetone and hexane (1:1) to afford a white solid, 118 mg, m.p. 132-134° C., EIMS M+ m/z 626, IR cm−1 (KBr), 2990, 2930, 2900, 2830 (OMe); 1675 (flavanone CO), 1600, 1570 and 1515 cm−1 (arom.); H—NMR &dgr;(CDCl3): 2.72 (2H, m, H-3″), 3.90 (6H, s, 233 OMe), 3.83 (6H, s, 2×OMe), 3.90 (6H, s, 2×OMe), 4.70 (1H, d, J=12 Hz, 3-H), 5.28 (1H, m, H-2″), 5.73 (1H, d, J=12 Hz, H-2), 6.08 (1H, d J=2 Hz, H-6), 6.15 (1H, s, H-6″), 6.17 (1H, d, J=2 Hz, H-8), 6.82 (2H, d, J=8 Hz, H-3′, 5′), 6.90 (2H, d, J=8 Hz, H-3″′, 5″′), 7.28 (2H, d, J=8 Hz, H-2′, 6′), 7.32 (2H, d, J=8 Hz, H-2″′, 6″′).

[0075] Robustaflavone hexamethyl ether. A solution of robustaflavone (200 mg, 0.272 mnol) in acetone (20 niL) and dimethylsulfate (2 mL) containing 3.8 g potassium carbonate was heated at reflux for 72 h with magnetic stirring. Filtration followed by evaporation of the filtrate afforded a black, oily semi-solid. Purification via column chromatography (silica gel, CHCl3/MeOH, 98:2) provided the desired product as an off-white solid (140 mg, 60.6%). Physical and spectral properties were identical to those previously reported in the literature.55

EXAMPLE 4 General Procedure for Preparation of Biflavanoid Sulfates

[0076] The dicyclohexylcarbodiimide (DDC)-mediated esterification of flavones and flavonols with tetrabutylammonium hydrogen sulfate (TBSHS) resulted in the formation of mono-, di-, and trisulfated products by controlling the reaction temperature and amount of reagents. Sulfation occurred mainly at positions 7,4′ and 3 of the flavonoid skeleton and followed the order 7>4′>3.80 Biflavanoid partial sulfate esters are prepared by treating the biflavanoid with TBAHS (tetrabutylammonium hydrogen sulfate) and DDC (dicyclohexylcarbodiimide) in pyridine using controlled amounts of reagents and temperature. The reaction product, sulfate ester TBA-salt, is separated from minor by-products by gel filtration. The sulfate ester TBA-salt is converted to the potassium salt by treatment with saturated methanolic potassium carbonate. The resulting potassium salt is purified by repeated chromatography on Sephadex G-10 column using a 0-50% gradient of aqueous methanol.&lgr;

Robustaflavone Tetrasulfate K-salt

[0077] A solution of robustaflavone (46.1 mg, 0.086 mM, 1.0 equivalent) in pyridine (5 mL) was treated with 1,3-dicyclohexylcarbodiimide (DCC) (500 mg, 2.423 mM, 28.17 equivalent) and tetrabutylammonium hydrogen sulfate (TBAHS) (97.5 mg, 0.287 mM, 3.34 equivalent) at 4° C. (in refrigerator) for 86 hours. The reaction solution was diluted with MEOH and the dicyclohexylurea precipitate was removed by filtration. The supernatant was chromatographed on Sephadex LH-20 (3 g, in MEOH) and eluted with MEOH and a MeOH-acetone (1:1) mixture. The yellow fractions containing robustaflavone tetrasulfate were concentrated to 5 mL and then treated with 15 mL of saturated K2CO3 in MEOH. The precipitate of robustaflavone tetrasulfate K-salt was collected by filtration and washed with MEOH (3ml×9) and water 3 mL×5), successively. The MEOH and water washes were collected separately. The water solution was lyophilized to obtain 72 mg robustaflavone-7,4′,7″,4″′-tetrasulfate K-salt as a yellow powder, 1H—NMR (DMSO, 300 MHz) &dgr;6.56 (1H, bs, H-6), 7.19 (1H, bs, H-8), 6.78 (1H, s, H-3), 7.75 (1H, dd, J=9.0, 2.0 Hz, H-6′), 7.87 (1H, d, J=9.0 Hz, H-5′), 8.31 (1H, d, J=2.0 Hz, H-2′), 6.85 (1H, s, H-8), 6.75 (1H, s, H-3″), 7.33 (2H, d, J=9.0 Hz, H-3″′, 5″′), 7.94 (2H, d, J=9.0 Hz, H-2″′, 6″′).

EXAMPLE 5 General Procedure for Preparation of Biflavanoid Acid Salt

[0078] The dried mixture of biflavanoid, appropriate acid anhydride, and appropriate catalyst, such as 4-dimethylaminopyridine are dissolved in dry pyridine. The solution is worked-up by standard methods to yield biflavanoid acid adduct. The biflavanoid acid can be converted to the potassium salt by treatment with saturated methanolic potassium carbonate.79

Robustaflavone Tetrasodium Salt

[0079] Robustaflavone (53.8 mg, 0.10 mmol) was dissolved in 0.400 mL of 1.0 M NaOH, and the resulting dark yellow solution was freeze dried. Following drying, 62 mg (99% of theoretical yield) of a brick-orange glass was obtained. The product is believed a mixture of salt forms which may include the tetrasodium salt form. Robustaflavone tetrasodium salt was assayed for activity and was approximately 10-fold less active than robustaflavone in the vitro assay against HBV (see Example 6). It was also moderately active against adenovirus type 1:EC50 =32 uM; IC50>160 uM.

EXAMPLE 6 Antiviral HBV Activity of Biflavanoids

[0080] In this example, robustaflavone and related biflavanoids were screened for hepatitis B (HBV) antiviral and cytotoxicity activity.

[0081] Antiviral HBV Assay. The inhibition of HBV replication in cultures of 2.2.15 cells was assayed using chronically HBV-producing human liver cells which were seeded into 24-well tissue culture plates and grown to confluence. Test compounds were added daily for a nine continuous day period; the culture medium was collected and stored for analysis of extracellular (virion) HBV DNA after 0, 3, 6, and 9 days of treatment. The treated cells were lysed 24 hours following day 9 of treatment for the analysis of intracellular HBV genomic forms. The overall levels of HBV DNA (both extracellular and intracellular DNA) and the relative rate of HBV replication (intracellular DNA) were analyzed quantitatively. The analysis was performed using blot hybridization techniques and [32P]-labeled HBV-specific probes. The HBV DNA levels were measured by comparison to known amounts of HBV DNA standards applied to every nitrocellulose membrane (gel or slot blot). An AMBIS beta scanner, which measures the radioactive decay of the hybridized probes directly from the nitrocellulose membranes, was used for the quantitative analysis. Standard curves, generated by multiple analyses, were used to correlate CPM measurements made by the beta scanner with relative levels of target HBV DNA. The levels of HBV virion DNA released into the culture medium were analyzed by a slot blot hybridization procedure. HBV DNA levels were then compared to those at day 0 to determine the effect of the test compound. A known positive drug was evaluated in parallel with test compounds in each test. This drug was 2′,3′-dideoxycytosine (2′,3′-ddC) or lamivdine (3TC). The data were expressed as 50% effective (virus-inhibitory) concentrations (EC50). The 90% effective concentration (EC90), which is that test drug concentration that inhibits virus yield by 1 log10, was determined from these data. Each test compound's antiviral activity was expressed as a selectivity index (SI), which is the CC50 or CC90, the concentration of compound which killed 50% or 90% of the treated cells, divided by the EC50. Generally an SI of 10 or greater is indicative of positive antiviral activity, although other factors, such as a low SI for the positive control, are also taken into consideration.

[0082] HBV Cytotoxicity Assays. The toxicity of the test compounds in cultures of 2.2.15 cells, grown to confluence in 96-well flat-bottomed tissue culture plates and treated with compounds as described above, were assayed at four concentrations each in triplicate cultures, in 3 to 10-fold steps. Untreated control cultures were maintained on each plate. On each plate, wells containing no cells were used to correct for light scattering. The toxicity was determined by the inhibition of the uptake of neutral red dye, determined by absorbance at 510 nm relative to untreated cells, 24 hours following day 9 of treatment.

[0083] Analysis of HBV Nucleic Acids and Proteins. HBV viron DNA in culture medium, and intracellular HBV RI and HBV RNA levels were determined by quantitative blot hydridization analyses (dot, Southern, and Northern blot, respectively).81,82 Nucleic acids were prepared by previously described procedures. Integrated HBV DNA, which remains at a stable level per cell during the treatment periods was used to quantitate the amount of cellular DNA transferred in each Southern gel lane.81,82 For the HBV RNA analyses, the levels of b-actin RNA were used to quantitate the amount of cellular RNA transferred in each Northern gel lane. Previous examinations of b-actin-specific RNA in confluent cultures of 2.2.15 cells demonstrated a steady state level of approximately 1.0 pg b-actin RNA/mg unfractionated cellular RNA.81 EC90 values (10-fold depression of HBV DNA levels relative to untreated control cultures) were determined by linear regression. 82 EC90 values were used for comparison since, in this culture system, DNA levels within 3-fold of control values are not generally statistically significant.83

[0084] Values of HBV proteins were determined by semi-quantitative EIA performed as previously described.83 For the EIA analyses, test samples were diluted (2- to 10-fold) so that the assay values produced were within the linear dynamic range of the EIA assays. Standard curves using serial dilutions of positive assay controls were included in each set of EIA analyses. HBV surface antigen (HBcAg), preSI protein, and HBc antigen (HBcAg) are released as extracellular products and were therefore analyzed in culture medium obtained 24 h following the last treatment dose of oligonucleotides or 2′, 3′-ddc. HBV core antigen (HBcAg) is an intracellular viral protein and was assayed in cell extracts produced by Triton-X-100 lysis.83

[0085] Cultures for HBV RNA were maintained on 6-well plates, cultures for HBV virion DNA analyses were maintained on either 96- or 24-well plates, and cultures for all other HBV parameters were maintained on 24-well plates.

[0086] The concentrations of antiviral agents used in these studies approximates the EC50 values of the individual agents against intracellular HBV DNA replication intermediates (HBV RI). Cultures were treated with the indicated agents for 9 days using standard procedures. Values reported are the levels of the indicated HBV markers at the end of the treatment period (“DAY 9”) expressed as a percentage (±standard deviation (S.D.) of the average levels in the control cultures at the beginning of the treatment period (“DAY 0”). The method of expression permits an analysis of the variation of the HBV markers in the untreated (control) cultures over the course of the treatment period. HBV nucleic acid levels were measured by standard blot hydridization (dot, Southern, or Northern). HBV protein levels were measured by standard semi-quantitative ELA methods. Cultures for HBV RNA were maintained in 6-well culture plates. The levels of each of two major classes of HBV RNA transcripts are listed separately. Cultures for all other HBV markers were maintained in 24-well culture plates. For each treatment, a total of 4 separate cultures were used for the analysis of each HBV marker at both DAY 0 and DAY 9.

[0087] Results. Tables 2 and 3 present evidence that robustaflavone is an extremely effective anti-HBV agent against the hepatitis B virus in comparison to the control drug, 2′,3′-ddC. It was observed from the results that robustaflavone exhibited an impressive in vitro activity against extracellular (virion) HBV DNA, with an effective average concentration (EC50) of 0.25 uM and an average selectivity index (CC50/EC90) of 153; compared to an effective average EC50 of 1.4 uM and average SI of 31 for 2′,3′-ddC. Furthermore, measurement of the relative rate of HBV replication intermediates (RI) (intracellular DNA) again indicates the effectiveness of robustaflavone over the control drug, 2′,3′-ddC. Robustaflavone exhibits an effective EC50 of 0.6 uM and SI of 80; compared to an EC50 of 2.4 uM and SI of 24 for 2′,3′-ddC. Volkensiflavone hexamethyl ether (6), rhusflavanone acetate (10) and succendaneaflavanone hexaacetate (12) exhibited moderate anti-HBV activity while amentoflavone (1), agathistflavone, hinokiflavone (4), volkensiflavone (5), rhusflavanone (9) and succendaneaflavanone possessed little or no anti-HBV activity.

[0088] In summary, measurement of the overall levels of HBV DNA (both extracellular and intracellular DNA) and the relative rate of HBV replication intermediate (RI) (intracellular DNA) clearly demonstrates the effectiveness of robustaflavone against HBV. 2 TABLE 2 Anti- HBV activity of biflavanoids, biflavanones, and related semi-synthetic derivatives Hepatitis B Virus (HBV) HBV Virion EC501 EC902 SI3 Sample &mgr;M &mgr;M (CC50/EC90) 2′,3′-ddC* 1.8 9.4 28 Lamivudine (3TC) 0.038 0.16 11200 Penciclovir 0.19 0.92 471 Robustaflavone 0.25 2.2(5.6) 153(60) (0.60) Robustaflavone 2.9 28 >36 hexamethyl ether Robustaflavone 0.73 2.8(11) >360(163) Hexaacetate (3.9) Amentoflavone (1) >100 >100 ND Agathisflavone (2) >100 >100 ND Robustaflavone (3) 0.25 2.4 153 Hinokiflavone (4) >100 >100 ND Volkensiflavone (5) >100 >100 ND Volkensiflavone 11 108 13 Hexamethylether (6) Rhusflavanone (9) >100 >100 ND Rhusflavanone 7.1 6.2 2.8 hexaacetate (10) Succedaneaflavanone (11) >100 >100 ND Succedaneaflavanone 3.5 128 1.9 hexaacetate (12) Robustaflavone sodium 3.1 12 88 salt Robustaflavone 0.4 3.6 110 tetrasulfate (17) *Positive drug control; *150% effective dose; *290% effective dose (EC90); *3selective index: CC50/EC90

[0089] 3 TABLE 3 Effect of Antiviral Agents on HBV Proteins and Nucleic Acids in 2.2.15 Cells Relative Levels of HBV Proteinsa and Nucleic Acids (Day 9, % of Day 0 Control ± SD) Viral DNA Viral mRNA Extra- Intra- 3.6 kb and 3.6 kb and cellular cellular 2.1 kb 2.1 kb Viral Antigens Treatment DNA DNA mRNA mRNA HBsAg HBeAg HbcAG Untreated 127 ± 8  103 ± 11  90 ± 12 101 ± 10 117 ± 11 108 ± 5   86 ± 10 cells 2′,3′-ddC 1 + 1 6 + 1 94 + 7  87 + 9  90 − 12  88 + 10 91 + 9 Robusta- 1 ± 1 5 ± 1 93 ± 10 106 ± 11 97 ± 6 86 ± 6 138 ± 8  flavone @ 10 &mgr;M aNucleic acid and protein levels were determined following nine days of continuous exposure of confluent cultures of 2.2.15 cells to robustaflavone, ddC, and drug-free media, and are relative to the levels on day zero, before addition of drug.

EXAMPLE 7 Antivirial HBV Activity of Combinations of Robustaflavone, 3TC and PCV

[0090] Lamivudine and penciclovir were purchased from Moravek Biochemical (La Brea, Calif., U.S.A.). Robustaflavone was isolated from the seeds of Rhus succedanea, as described above.

[0091] Assay of anti-HBV activity and toxicity in 2.2.15 cells. Determination of therapeutic concentrations (EC50 and EC90) and toxicity concentrations (IC50) were determined as previously described (90). Stock solutions of text drugs were prepared in DMSO. Culture medium was changed daily and analyzed for extracellular HBV DNA after nine days of continuous drug exposure. Extracellular HBV DNA was extracted from the culture media and analyzed by a slot blot hydridization technique, using a 32P-labeled Eco RI HBV DNA fragment, as previously described (91), and quantitated via comparison to HBV standards on a nitrocellulose filter, using an AMBIS beta scanner. Toxicity was determined by measuring inhibition of neutral red dye uptake. Effective concentration (EC50 and EC90) and toxicity (IC50) values were calculated via comparison with drug-free controls. Effective concentration values for individual agents are the mean of six cultures per concentration point. Toxicity values are the mean of three cultures per concentration point.

[0092] Combination Studies of Robustaflavone, 3TC and PCV in 2.2.15 cells. Cultures were treated with combination of agents as previously described (92). Briefly, antiviral agents were mixed at approximately equipotent molar concentrations based on the EC50 values. Serial dilutions of these mixtures were then used to treat cultures as described above along with the appropriate monotherapies. For these studies, eight cultures were used for each of six 3-fold dilutions.

[0093] Determination of Mechanism of Action. Measurements of intracellular and extracellular HBV DNA, 3.6 kb and 2.1 kb mRNA fragments, and HBsAg, HBeAg and HBcAg protein markers were conducted as previously described (93).

[0094] Results and Discussion. Robustaflavone exhibited activity against HBV replication in a chronically infected (90) human hepatoblastoma cell line (2.2.15), inhibiting the replication of HBV by 50% relative to drug-free controls at a concentration of 0.25 &mgr;M (EC50, mean of two trials), with an in vitro therapeutic index (TI, IC50/EC90) of 153 (Table 2). None of the other compounds tested from the series of biflavanoids showed significant activity against HBV with an acceptable selectivity index (Table 2); thus, robustaflavone was the only derivative selected for further evaluation. In a comparison with several nucleoside antiviral agents, the anti-HBV activity of robustaflavone was superior to ddC (EC50=1.4 &mgr;M, TI=30) and similar to penciclovir (EC50=0.19 &mgr;M, TI=471). Lamivudine was clearly the most active of the agents evaluated (EC50=0.038 &mgr;M, TI=11200).

[0095] Combinations of antiviral agents are now accepted to be superior to monotherapy in certain instances, particularly in treatment of HIV infection, because of the ability of drug combinations to overcome resistant mutants which accumulate following exposure to a single agent (89). Clinical isolates of 3TC-resistant mutants of HBV have recently been reported following monotherapy with that agent (87,88).

[0096] Robustaflavone was evaluated in combination with two leading anti-HBV candidates, 3TC and penciclovir (the bioactive form of famciclovir), in an effort to determine if combinations of robustaflavone with either of these drugs could potentially offer synergistic benefits if used as part of an anti-HBV regimen. As shown in Table 4, combinations of robustaflavone (RF) with 3TC at differing ratios showed varying degrees of syngergism and antagonism. It is interesting to note that the most effective ratio of RF to 3TC was 10:1, which exhibited an EC50 of 0.054 &mgr;M with respect to RF (0.0054 &mgr;M with respect to 3TC). The ratio of 10:1 correlates well with the EC50 ratio for RF and 3TC (RF/3TC=6.6). The concentration of 3TC in the 10:1 combination (0.038 &mgr;M) necessary to achieve 90% reduction of HBV replication was lower than that required by 3TC alone (0.16 &mgr;M), illustrating the potential benefits of the combination. Ratios of RF:30:1 and 3:1 were less effective than the 10:1 conbination. Combostat® analysis96 of the RF/3Tccombination data indicated that, at a molar ratio of 10:1, the combination exhibited syngergism at all dilutions. The RC:3TC ratio of 30:1 exhibited antagonism at lower drug concentrations, and additive or mildly syngergistic effects at higher concentrations. At a RF:3TC ratio of 3:1, the combination exhibited antagonism at all drug concentrations.

[0097] The combinations of RF with penciclovir (PCV) also showed a clear syngergistic effect. Because RF and PCV have similar effective concentrations (EC50 values of 0.25 and 0.19 &mgr;M, respectively) and cellular toxicity concentrations (IC50 values of 337 and 433 &mgr;M, respectively), the synergistic effects of the agents together was much more obvious. At a RF:PCV ratio of 10:1, the selectiveity index was inferior to either drug alone. At a RF:PCV ratio of 3:1, the selectivity index improved to 570, higher than either drug alone, while a RF:PCV ratio of 1:1 exhibited an even higher selectivity index of 684. At the 1:1 combination ratio, the concentration of PCV necessary to achieve 90% reduction of HBV replication was 0.51 &mgr;M, nearly two-fold less than the concentration required of PCV alone (0.92 &mgr;M). Combostat® analysis of the combination data indicated that a RF:PCV ratio of 1:1 exhibited synergism at all concentrations. At a RF:PCV ratio of 3:1, the combination still showed a mild degree of syngergism at all concentrations except for the very lowest, while a 10:1 ratio exhibited antagonism at most concentrations. Again, the ratio exhibiting the greatest syngergistic effect was that which mirrored the EC50 ratio for the two agents (RF/PCV=1.3).

[0098] To determine the likely mechanism of action for robustaflavone, levels of extracellular and intracellular HBV DNA, mRNA and protein antigen markers were measured in 2.2.15 cells on day nine following continuous treatment (93). As presented in Table 3, the levels of extracellular and intracellular HBV DNA were dramatically decreased relative to drug-free controls for both robustaflavone and ddC, which was used as a positive control. Neither the levels of viral mRNA (3.6 and 2.1 kb) nor the three major viral antigens (HBsAg, HBeAg and HBcAg) were significantly affected by exposure of the cells to robustaflavone. These results strongly suggest that robustaflavone acts via inhibition of the viral DNA polymerase, supported by the fact that results for robustaflavone were essentially identical to those for ddC, an established inhibitor of viral polymerases, used as a positive control.

[0099] In conclusion, robustaflavone represents a novel non-nucleoside natural product possessing impressive activity against hepatitis B virus replication, determined in a chronically replicating transfected human cell line. The paucity of agents available for the treatment of HBV infection underscore the need for development of new lead compounds, especially non-nucleoside structures, which would complement the drugs currently in development. 4 TABLE 4 Antiviral and cytotoxicity effects of robustaflavone (RF), lamivudine (3TC), penciclovir (PCV) and drug combinations in 2.2.15 cellsa Drug EC50 EC90 IC50 TI 3TC or (or combination) (&mgr;M)b (&mgr;M)b (&mgr;M) (IC50/EC90) PCV (&mgr;M)c 3TC 0.038 0.16 1790 11200 — PCV 0.19 0.92 433 471 — RF 0.25 2.2 337 153 — RF + 3TC (30:1) 0.33 1.7 325 191 0.057 RP + 3TC (10:1) 0.054 0.38 340 894 0.038 RF + 3TC (3:1) 0.16 0.73 363 497 0.24 RF + PCV (10:1) 0.99 2.9 334 115 0.29 RF + PCV (3:1) 0.22 0.63 360 570 0.21 RF + PCV (1:1) 0.11 0.51 349 684 0.51 aSingle agents studies were conducted as described in reference 90. Combination studies were conducted as described in reference 92. bEffective concentration necessary to decreased HBV DNA levels by 50% (EC50) or 90% (EC90) relative to drug-free controls. cConcentration of 3TC or PCV in combination with RF required to achieve inhibition of HBV replication by 90% (EC90)

EXAMPLE 8 Anti-Respiratory Viral Activity of Biflavanoids

[0100] In this example, robustaflavone and related biflavanoids were screened for respiratory (influenza A and B, RSV, parainfluenza 3, adenovirus 5, and measles) antiviral and cytotoxic activities.

[0101] Anti-Respiratory Viral Assay. The viruses used in the primary screen for antiviral activity against respiratory viruses consisted of: (1) Influenza A and B—Virus strains: A/Texas/36/91 (H1N1) (Source: Center for Disease Control (CDC), A/Beijing/2/92 (H3N2) (Source: CDC), B/Panama/45/90 (Source: CDC), A/NWS/33 (H1N1) (Source: American Type Culture Collection [ATCC]). (All but A/NWS/33 are tested in the presence of trypsin.); cell lines: Madin Darby canine kidney (MDCK) cells; (2) Respiratory syncytial virus—Virus strain: Utah 89 (Source: Utah State Diagnostic Laboratory, cell line: African green monkey kidney (MA-104) cells; (3) Parainfluenza type 3 virus—Virus stain: C243 (Source: ATCC); cell line: African green monkey kidney (MA-104) cells; (4) Measles virus—Virus strain: CC (Source: Pennsylvania State University; cell line: African green monkey kidney (BSC-1) cells; and (5) Adenovirus type 5—Virus strain: Adenoid 75 (Source: ATCC); cell line: Human lung carcinoma (A549) cells.

[0102] Test compounds were assayed for continual activity and cytotoxicity. Three methods were used for assay of antiviral activity: (1) inhibition of the viral cytopathic effect (CPE); (2) increase in the neutral red (NR) dye uptake; and (3) decrease in the virus yield. Methods for ascertaining cytotoxicity were visual observation, neutral red uptake, and viable cell count.32

[0103] Inhibition of the Viral Cytopathic Effect (CPE). The test for CPE was run in 96-well flat-bottomed microplates and was used for the initial antiviral evaluation of all new test compounds. In this CPE inhibition test, seven one-half log10 dilutions of each test compound were added to 4 cups containing the cell monolayer; within 5 min, the virus was then added and the plate sealed, incubated at 37° C. and CPE read microscopically when untreated infected controls develop a 3 to 4+ CPE (approximately 72 h). A known positive drug was evaluated in parallel with test drugs in each test. This drug was ribavirin for influenza, measles, respiratory syncytial, and parainfluenza viruses, and (S)-1-(3-hydroxy-2-phosophonylmethoxypropyl)adenine (HPMPA) for adenovirus. The data were expressed as 50% effective (virus-inhibitory) concentrations (EC50).

[0104] Increase in the Neutral Red (NR) Dye Uptake. The test for increase in the NR dye uptake was run to validate the CPE inhibition seen in the initial test, and utilizes the same 96-well microplates after the CPE has been read. Neutral red dye was added to the medium; cells not damaged by virus take up a greater amount of dye, which was read on a computerized microplate autoreader. An EC50 value was determined from this dye uptake.

[0105] Decrease in virus yield. Compounds considered active by CPE inhibition and NR uptake were retested using both CPE inhibition, and, using the same plate, the effect on reduction of virus yield was determined by assaying frozen and thawed eluates from each cup for virus titer by serial dilution onto monolayers of susceptible cells. Development of CPE in these cells was an indication of presence of infectious virus. As in the initial tests, a known active drug (ribavirin) was run in parallel as a positive control. The 90% effective concentration (EC90), which was that test drug concentration that inhibits virus yield by 1 log10, was determined from these data.

[0106] Cytotoxicity Assays. These assays consist of visual observation, neutral red dye uptake, and viable cell count.

[0107] Visual Observation—In the CPE inhibition tests, two wells of uninfected cells treated with each concentration of test compound were run in parallel with the infected, treated wells. At the same time CPE was determined microscopically, the toxicity control cells were examined microscopically for any changes in cell appearance compared to normal control cells run in the same plate. These changes were given a designation conforming to the degree of cytotoxicity seen (e.g., enlargement, granularity, cells with ragged edges, a cloudy appearance, rounding, detachment from the surface of the well, or other changes. These changes were given a designation of T (100% toxic), Pvh (partially toxic-very heavy 80%), Ph (partially toxic-heavy 60%), P (partially toxic-40%), Psi partially toxic-slight-20%), or 0 (no toxicity −0%), conforming to the degree of cytotoxicity seen. A 50% cell inhibitory (cytotoxic) concentration (IC50) was determined by regression analysis of the data.

[0108] Neutral Red Dye Uptake—In the neutral red dye uptake phase of the antiviral test described above, the two toxicity control wells also receive neutral red dye and the degree of color intensity was determined spectrophotometrically. A neutral red IC50 was subsequently deternmined.

[0109] Viable Cell Count—Compounds considered to have significant antiviral activity in the initial CPE and NR tests were retested for their effects on cell growth. In this test, 12-well tissue culture plates were seeded with cells (sufficient to be approximately 20% confluent in the well) and exposed to varying concentrations of the test drug while the cells were dividing rapidly. The plates were then incubated in a CO2 incubator at 37° C. for 72 h, at which time the media-drug solution was removed and the cells washed. Trypsin was added to remove the cells, which were then counted using a Coulter cell counter. An IC50 was then determined using the average of three separate counts at each drug dilution.

[0110] Each test compound's antiviral activity was expressed as a selectivity index (SI), which was the IC50 or IC90 divided by EC50. Generally an SI of 10 or greater was indicative of positive antiviral activity, although other factors, such as a low SI for the positive control, were also taken into consideration.

Anti-Influenza A and Anti-Influenza B Activity

[0111] Compounds 1-6 and 9-12 have been screened for inhibitory activity against influenza A (strains H1N1 and H3N2) and influenza B viruses. For these compounds both cytopathic effect inhibition (CPE) and neutral red uptake test methods were investigated. The results are displayed on Tables 5-7. For the results shown in Tables 5-7 the selective index (SI) is calculated as IC50 (50% cell inhibitor (cytotoxic) concentration) over the EC50 (50% effective concentration).

[0112] Influenza A. Tables 5 and 6 provide data that robustaflavone (3) had significant antiviral activity towards two influenza A strains, when compared to the control drug, ribavirin. The effective concentrations (EC50) of robustaflavone (3) were 1.9 &mgr;g/mL for both influenza A H1N1(Table 5) and H3N2 (Table 6) strains, as compared to 1.9 and 4.1 ug/mL for the control drug, ribavirin. The IC50 values for robustaflavone were 18 and 32 ug/mL, respectively for H1N1 and H3N2 in the CPE assay. However, the selectivity indexes (SI) for ribavirin were 296 and 137 against influenza A strains H1N1 and H3N2, respectively, as compared to 9.5 and 17 for robustaflavone (3). The effective neutral red concentrations (EC50) of robustaflavone (3) were 2.0 and 1.8 ug/mL for influenza A strains H1N1 and H3N2, respectively and the IC50 values were ˜32 and ˜100 ug/miL. This compared favorably with ribavirin, which had effective neutral red concentrations of 1.4 and 5.7 ug/mL, respectively for these strains. The SI's for neutral red uptake for ribavirin were 132 and 70, respectively, toward influenza A strains H1N1 and H3N2, whereas those for robustaflavone (3) were 16 and 56.

[0113] Amentoflavone (1) also demonstrated significant antiviral activity against both strains of influenza A. The EC50 values of amentoflavone (1) were 3.1 and 4.3 &mgr;g/mL, respectively in CPE inhibition tests. The IC50 values were 22 and >100 &mgr;g/mL, therefore it had SI values of 7.1 and >23 for influenza A strains H1N1 and H3N2. The other biflavanoids assayed were either inactive or toxic, except for agathisflavone which produced an SI value of >18 for the neutral red assay, but only 1 for the CPE assay. The acetylation of rhusflavanone (9), to rhusflavanone hexaacetate (10), slightly increased both the activity and toxicity against both influenza A strains in both assays. The acetylation of succedaneaflavanone (11) did not change the activity or toxicity considerably, and methylation of volkensiflavone (5) to volkensiflavone hexamethyl ether (6) resulted in a decrease in both the activity and the toxicity, in both the CPE inhibition and the neutral red assays. As shown in Tables 5 and 6, the modifications to these three compounds did result in changes in activity and toxicity, but none produced significant changes in the SI value. 5 TABLE 5 Influenza A (H1N1) Virus: Texas/36/91 CPE Inhibition Neutral Red EC50*1 IC50*2 EC50*1 IC50*2 Sample ug/mL ug/mL SI*3 ug/mL ug/mL SI*3 Ribavirin* 1.9 562 296 1.4 185 132 Amentoflavone (1) 3.1 22 7.1 5.3 >100 19 Agathisflavone (2) 6.6 6.5 1.0 5.6 >100 18 Robustaflavone (3) 1.9 18 9.5 2.0 32 16 Hinokiflavone (4) >1.0 1.4 <1.4 1.8 2.0 1.1 Volkensiflavone (5) >32 13 0 15 14 1.0 Volkensiflavone ˜100 <24 0 ˜100 ˜100 0 hexamethyl ether (6) Rhusflavanone (9) >10 8.2 0 24 26 1.1 Rhusflavanone >10 7.2 0 5.6 5.7 1.0 hexaacetate (10) Succedanea- >3.2 4.9 <1.5 5.2 5.0 1.0 flavanone (11) Succedanea- 5.6 8.2 1.5 7.4 7.4 1.0 flavanone hexaacetate (12) *Positive control drug; *150% effective dose; *250% cell inhibitory (cytotoxic) concentration; *3selective index: IC50/EC50

[0114] 6 TABLE 6 Influenza A (H3N2) Virus: Beijing/32/92 CPE Inhibition Neutral Red EC50*1 IC50*2 EC50*1 IC50*2 Sample ug/mL ug/mL SI*3 ug/mL ug/mL SI*3 Ribavirin* 4.1 562 137 5.7 397 70 Amentoflavone (1) 4.3 >100 >23 6.5 >100 >15 Agathisflavone (2) 24 18 0.8 13 19 1.5 Robustaflavone (3) 1.9 ˜32 17 1.8 ˜100 56 Hinokiflavone (4) >3.2 1.3 0 1.9 2.2 1.2 Volkensiflavone (5) 56 42 0.8 38 37 1.0 Volkensiflavone ˜100 ˜100 0 ˜100 ˜100 0 hexamethyl ether (6) Rhusflavanone (9) >32 24 0 31 31 1.0 Rhusflavanone >10 5.6 0 5.4 5.3 1.0 hexaacetate (10) Succedanea- >10 12 <1.2 12 12 1.0 flavone (11) Succedanea- 8.8 12 1.4 5.6 5.6 1.0 flavone (12) *Positive control drug; *150% effective dose; *250% cell inhibitory (cytotoxic) concentration; *3selective index: IC50/EC50

[0115] Influenza B. Table 7 indicates that robustaflavone had significant antiviral activity towards influenza B, when compared to the control drug, ribavirin. The effective concentration (EC50) of robustaflavone was an impressive 0.23 ug/mL, compared to 1.5 for ribavirin. The selectivity index (SI) for ribavirin was >667 against influenza B; as compared to <435 for robustaflavone. The effective neutral red concentration (EC50) of robustaflavone was 0.22 ug/mL, compared to the control drug, ribavirin, 0.48 ug/mL. The SI for neutral red uptake for ribavirin was 208, compared to 454 for robustaflavone. 7 TABLE 7 Influenza B Virus: Panama/45/90 CPE Inhibition Neutral Red EC50*1 IC50*2 EC50*1 IC50*2 Sample ug/mL ug/mL SI*3 ug/mL ug/mL SI*3 Ribavirin* 1.5 >1000 >667 0.48 100 208 Amentoflavone 0.56 100 178 — — — (1) Agathisflavone 3.2 18 5.6 — — — (2) Robustaflavone 0.23 ˜100 ˜435 0.22 ˜100 454 (3) Hinokiflavone (4) >1.0 1.2 <1.2 1.9 2.0 1.0 Volkensiflavone 1.1 38 34 4.5 20 4.4 (5) Volkensiflavone 2.6 ˜100 ˜38 <20 ˜100 5.0 hexamethyl ether (6) Rhusflavanone >4.1 38 9.3 — — — (9) Rhusflavanone >10 4.2 0 — — — hexaacetate (10) Succedanea- 0.97 15 15 2.2 7.0 3.2 flavanone (11) Succedanea- 5.4 12 2.2 5.9 5.9 1.0 flavanone hexaacetate (12) *Positive control drug; *150% effective dose; *250% cell inhibitory (cytotoxic) concentration; *3selective index: IC50/EC50

[0116] Amentoflavone (1) (I-3′-II-8 biapigenin), volkensiflavone (5) (naringenin I-3-II-8 apigenin), volkensiflavone hexamethyl ether and succedaneaflavanone (11) (I-6-II-6 binaringenin) also exhibited favorable antiviral activity against influenza B, having SI values of 178, 34, 38, and 15, respectively in the CPE assay. Agathisflavone (2)(I-6-11-8 biapigenin) and rhusflavanone (9) (I-6-II-8 binaringenin) demonstrated activity against influenza B virus, with SI values of 5.6 and 9.3, for the CPE assay. However in neutral red uptake tests, these biflavanoids showed no significant activity. None of the other biflavinoids assayed contributed significant activity. Methylation of volkensiflavone (5), to volkensiflavone hexamethyl ether (6) led to lower activity and decreased cytotoxicity.

[0117] All of these biflavanoids were relatively inactive toward parainfluenza type 3, respiratory syncytial, measles, and adenovirus type 5 viruses, as shown in Table 8 and Table 9, except amentoflavone (1) and rhusflavanone (9) which exhibited some slight activity against respiratory syncytial virus and measles virus, respectively. 8 TABLE 8 Measles Virus Adenovirus Type 5 CPI Inhibition Neutral Red CPE Inhibition Neutral Red EC50*1 IC50*2 EC50*1 IC50*2 EC50*1 IC50*2 EC50*1 IC50*2 Sample ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 Ribavirin* 3 150 50 1 150 150 — — — — — — HPMPA* — — — — — — 30 80 3 8 40 5 Amentoflavone (1) <40 <10 0 <20 <60 3 >100 18 0 >100 74 0 Agathisflavone (2) <60 <10 0 <10 <30 3 15 18 1 22 37 1 Robustaflavone (3) <14 <14 1 −30 ˜100 1 >100 56 0 >100 102 0 Hinokiflavone (4) <3 <4 1 <5 <11 2 >10 22 0 19 27 1 Volkensiflavone (5) <12 <10 1 <6 <10 1 56 47 1 >32 30 0 Volkensiflavone <70 <60 1 <7 <13 1 >100 47 0 >100 50 0 hexamethyl ether (6) Rhusflavanone (9) 14 21 2 5 40 8 56 47 1 33 15 0 Rhusflavanone ˜3 <4 0 <10 <10 0 >10 22 0 19 26 1 hexaacetate (10) Succedaneaflavone (11) ˜32 <23 0 <12 <20 1 10 22 0 19 27 1 Succedaneaflavone ˜3.2 <4 0 <2 <200 <1 20 19 1 6 6 1 hexaacetate (12) *Positive control drug; *150% effective dose; *250% cell inhibitory (cytotoxic) concentration, *3selective index: IC50/EC50

[0118] 9 TABLE 9 Parainfluenza Type 3 Virus Respiratory Syncytial Virus CPI Inhibition Neutral Red CPE Inhibition Neutral Red EC50*1 IC50*2 EC50*1 IC50*2 EC50*1 IC50*2 EC50*1 IC50*2 Sample ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 Ribavirin* 25 245 10 17 331 19 12 120 10 6 60 10 Amentoflavone (1) >100 ˜56 0 ˜32 ˜56 2 ˜10 ˜56 6 ˜21 ˜35 2 Agathisflavone (2) >100 ˜33 0 >100 ˜34 0 >10 ˜8 0 ˜24 ˜15 0 Robustaflavone (3) <100 56 0 39 79 2 >100 ˜56 0 ˜50 ˜100 2 Hinokiflavone (4) <1 1 0 5 7 1 >3.2 2.5 0 >3.2 6 0 Volkensiflavone (5) 47 50 1 15 44 3 32 56 2 28 13 0 Volkensiflavone <32 ˜15 0 >32 ˜33 0 ˜18 ˜30 2 >32 ˜59 >1 hexamethyl ether (6) Rhusflavanone (9) >100 47 0 85 30 0 >10 18 0 27 20 0 Rhusflavanone >10 22 0 32 19 0 >10 13 0 19 32 2 hexaacetate (10) Succedaneaflavone (11) >10 ˜22 0 23 ˜29 0 >10 ˜13 0 ˜16 ˜10 0 Succedaneaflavone >10 ˜14 0 >32 ˜13 0 >3 4 0 6 6 1 hexaacetate (12) *Positive control drug; *150% effective dose; *250% cell inhibitory (cytotoxic) concentration; *3selective index: IC50/EC50

EXAMPLE 9 Anti-HIV Viral Activity of Biflavanoids

[0119] We have investigated the anti-HIV-1 RT activity of biflavanoids isolated from Rhus succedanea, amentoflavone (1), agathisflavone (2), robustaflavone (3), hinokiflavone (4), rhusflavanone (9), succedaneaflavanone (11), and from Garcinia multiflora, volkensiflavone (5), morelloflavone (7), GB-1a (13), GB-1a 7″-O-b-glucoside (15), GB-2a (16), and their sulfate potassium salt, methyl ether, and acetyl derivative, volkensiflavone hexaacetate (6), morelloflavone heptamethyl ether (8), rhusflavanone hexaacetate (10), succedaneaflavanone hexaacetate (12), GB-1a hexamethyl ether (14), and robustaflavone tetrasulfate potassium salt (17).

[0120] Anti-HIV-1 RT Assay. The HIV-1 RT is a 66-kDa recombinant enzyme obtained from an Escherichia coli expression system using a genetically engineered plasmid; the enzyme was purified to near homogeneity. Synthetic DNA segments were used to introduce initiation and termination codons into the HIV-1 RT coding sequence, which permits expression of large quantities of HIV-1 RT in E. coli. The enzyme was shown to be active in RT assays and was inhibited by several known antiretroviral agents (e.g. AZT and suramin). The purified recombinant enzyme was sufficiently similar to the viral enzyme that it can be substituted for the latter in drug screening assays. The recombinant HIV-1 RT preparation used in all experiments had a protein concentration of 0.11 ug/mL and an activity of 238 nmol TTP incorporated per 10 min per mg of protein at 37° C. Prior to performing an experiment, the enzyme was diluted tenfold with buffer analogous to that used in the assay.

[0121] The assay mixture (final volume 100 uL) contained the following: 50 mM Tris-HCl buffer (pH 8.0), 150 mM KCl, 5 mM MgCl, 0.5 mM ethylene glycol-bis-(b-aminoethylether)-N,N′-tetraacetic acid (EGTA), 5 mM dithiothreitol 0.3 mM glutathione, 2.5 mg/mL bovine serum albumin, 41 mM poly A [S260 (mM)=7.8], 9.5 mM oligo (dT)12-18 [S265(mM)=5.6], 0.05% Triton X-100, 20 mM TTP, and 0.5 mCi of [3H]TTP. The reaction was started by the addition of 10 uL of HIV-1 RT, and the mixture was permitted to incubate at 37° C. for 1 h. Reactions were terminated by the addition of 25 uL of 0.1 M EGTA followed by chilling on ice. Aliquots of each reaction mixture (100 uL) were then spotted uniformly onto circular 2.5 cm DE-81 (Whatman) filters, kept at ambient temperature for 15 minutes, and washed four times with 5% aqueous Na2HPO47H2O. This was followed by two more washings with doubly distilled H2O. Finally, the filters were thoroughly dried and subjected to scintillation counting in a nonaqueous scintillation fluid.

[0122] For testing enzyme inhibition, five serial dilutions of samples in DMSO (10 uL) were added to the reaction mixtures prior to the addition of enzyme (10 uL). The final DMSO concentration used was 10%. The highest concentration of pure natural products and plant extracts tested was 200 &mgr;g/mL. Control assays are performed without the compounds or extracts, but an equivalent volume of DMSO was added. Fargaronine chloride was used as the positive control substance. This compound was isolated from Fagara xanthoxyloides Lam. Other positive control substances used were suramin (IC50 18 &mgr;g/mL) and daunomycin (IC50 125 &mgr;g/mL). The assay procedure and the concentration of all components were the same as that described above.47

[0123] Anti-HIV-1 RT Assay in Primary Human Lymphocytes

[0124] Cell Culture. Human PBM cells from healthy HIV-1 seronegative and hepatitis B virus seronegative donors were isolated by Ficoll-Hypaque discontinuous gradient centrifugation at 1,000 x g for 30 min, washed twice with phosphate-buffered saline (pH 7.2, PBS), and pelleted by centrifugation at 300 x g for 10 min. Before infection, the cells were stimulated by phytohemagglutinin (PHA) at a concentration of 6 &mgr;g/mL for 2-3 days in RPMI 1640 medium, supplemented with 15% heat-inactivated fetal calf serum, 1.5 mM L-glutamine, penicillin (100 U/mL), streptomycin (100 &mgr;g/mL), and 4 mM sodium bicarbonate buffer.

[0125] Viruses. HIV-1 (strain LAV-1) was obtained from Dr. P. Feorino (Emory University, Atlanta, Ga.). The virus was propagated in human PBM cells using RPMI 1640 medium, as described previously58 without PHA or fungizone and supplemented with 26 units/mL of recombinant interleukin-2 (Cetus Corporation, Emeryville, Calif.) and 7 &mgr;g/mL DEAE-dextran (Pharmacia, Uppsala, Sweden). Virus was obtained from cell-free culture supernatant and was titrated and stored in aliquots at −70° C. until use.

[0126] Inhibition of Virus Replication in Human PBM Cells. Uninfected PHA-stimulated human PBM cells were infected in bulk with a suitable dilution of virus. The mean reverse transcriptase (RT) activity of the inocula was about 60,000 dpm RT activity/106 cells/10 mL. This represents, by a limiting dilution method in PBM cells, a multiplicity of infection of about 0.01. After 1 h, the cells were uniformly distributed among 25 cm2 flasks to give a 5 mL suspension containing about 2×106 cells/mL each. The samples at twice their final concentration in 5 mL of RPMI 1640 medium, supplemented as described above, were added to the cultures. The cultures were maintained in a humidified 5% CO2-95% air incubator at 37° C. for six days after injection, at which point all cultures were sampled for supematant RT activity. Previous studies had indicated that maximum RT levels were obtained at that time.

[0127] RT Activity Assay. A volume of supernatant (1 mL) from each culture was clarified of cells at 300 x g for 10 min. Virus particles were pelleted at 12,000 rpm for 2 h using a Jouan refrigerated microcentrifuge (Model MR 1822) and suspended in 100 &mgr;L of virus disrupting buffer (50 mM Tris-HCl, pH 7.8, 800 mM NaCl, 20% glycerol, 0.5 mM phenylmethyl sulfonyl fluoride, and 0.5% Triton X-100).

[0128] The RT assay was performed in 96-well microtiter plates, as described by Spira.69 The reaction mixture, which contained 50 mM Tris-HCl, pH 7.8, 9 mM MgCl2, 5 mM dithiothreitol, 4.7 &mgr;g/mL (rA)n(dT)12-18, 140 &mgr;M dAPT, and 0.22 &mgr;M [3H]TTP (specific activity 78.0 Ci/mmol, equivalent to 17,300 cpm/pmol; NEN Research Products, Boston, Mass.), was added to each well. The sample (20 &mgr;L) was added to the reaction mixture, which was then incubated at 37° C. for 2 h. The reaction was terminated by the addition of 100 &mgr;L of 10% trichloroacetic acid (TCA) containing 0.45 mM sodium pyrophosphate. The acid-insoluble nucleic acids which precipitated were collected on glass filters using a Skatron semi-automatic harvester (setting 9). The filters were washed with 5% TCA and 70% ethanol, dried and placed in scintillation vials. Scintillation fluid (Ecolite, ICN, Irvine, Calif.) (4 mL) was added and the amount of radioactivity in each sample was determined using a Beckman liquid scintillation analyzer (Model LS 3801). The results were expressed in dpm/mL of original clarified supernatant. The procedures for the anti-HIV assays in PBM cells described above have been published.67,69

[0129] Cytotoxicity Studies in PBM Cells. The compounds were evaluated for their potential toxic effects on uninfected PHA-stimulated human PBM cells. The cells were cultured with and without drug for 24 h, at which time radiolabeled thymidine was added. The assay was performed as described previously.35 Alternately, cells are counted on day 6 using a hemacytometer and/or Coulter counter as described previously.68

[0130] Median-Effect Method. EC50 and IC50 values were obtained by analysis of the data using the median-effect equation.42 These values were derived from the computer-generated median effect plot of the dose-effect data using a commercially available program.43

[0131] The results shown in Table 10 indicate that both hinokiflavone (4) and robustaflavone (3) demonstrated similar activity against HIV-1 RT at an IC50 (50% inhibition dose) of 35.2 &mgr;g/mL and 33.7 &mgr;g/mL, respectively. The water soluble form of robustaflavone, robustaflavone tetrasulfate K-salt (17) exhibited 95.5% inhibition at a concentration of 200 ug/mL, with an IC50 value of 144.4 ug/nL. Amentoflavone (1), agathisflavone (2), morelloflavone (7), GB-1a (13), and GB-2a (16) were moderately active against HIV-1 RT with IC50 values of 64.0 &mgr;g/mL, 53.8 &mgr;g/mL, 64.7 &mgr;g/mL, 127.8 &mgr;g/mL, and 94.6 &mgr;g/mL, respectively. The other biflavanoids were either slightly active or inactive against HIV-1 RT.

[0132] The results of both studies are presented in Table 10. The results of the inhibitory activity tests using HIV-1 RT enzyme (p66/p51 heterodimer) indicated that the biflavones, two apigenin units linked either with C—C or C—O—C bonds, exhibited significant activity. Robustaflavone (3) (two apigenins linked through an I-6-II-3′ linkage) and hinokiflavone (4) (I-6-O-II-4′ linkage) demonstrated similar activity, with 50% inhibition (IC50) at doses of 35.2 &mgr;g/mL and 33.7 &mgr;g/mL, respectively. The IC50 values of amentoflavone (1) (I-8- II-3′linkage) and agathisflavone (2) (I-6-II-8 linkage) were 64.0 &mgr;g/mL and 53.8 &mgr;g/mL, respectively. 10 TABLE 10 Anti-HIV-1 RT Activity of Biflavanoids Anti-HIV-1 RT Ant-HIV-1 Cytotoxicity % Inhibition in PBM in PBM Selective at 200 IC50 cells cells Index Compounds ug/ml (uM) EC50 (uM) IC50 (uM) (SI) Apigenin 72 (443) Naringenin 34.9 Weakly Active Amentoflavone (1) 97.3 (119) >10.94 35 Agathisflavone (2) 99.8 (100) 7.3, 6.0 25 <1 Robustaflavone (3) 91.4  (65) >100 77 <1 Hinokiflavone (4) 89.0  (62) 4.1 9.1 <1 Volkensiflavone (5) 45.3 Weakly Active 2.2 Volkensiflavone 0.00 Inactive Me2 (6) Morelloflavone (7) 99.2 (116) 5.7, 8.0 82 12 Rhusflavanone (9) 14.1 Inactive Rhusflavanone Ac6 (10) 0.00 Inactive Succedanea- 22.1 Inactive flavanone (11) Succedanea- 0.00 Inactive flavanone Ac6 (12) GB-1a (13) 86.0 (236) 38.0 88 2.3 GB-1a Me6 (14) 0.00 Inactive GB-1a glucoside (15) 1.46 Inactive GB-2a (16) 96 (170) Robustaflavone 95.5 144.4 tetrasulfate K-salt

[0133] Biflavanoids constructed of flavanone-flavone units through I-3-II-8 linkages were moderately to weakly active, i.e. morelloflavone (7) (naringenin I-3-II-8 quercetin) demonstrated moderate activity, with an IC50 value of 64.7 &mgr;g/mL, while volkensiflavone (5)(narnigenin I-3-II-8 apigenin) was weakly active. Biflavanones consisting of two naringenin units or naringenin-eriodictol through I-3-II-8 linkages exhibited moderate activity, such as GB-1a (13) (IC50 127.8 &mgr;g/ML) and GB-2a (16) (IC50 94.6 &mgr;g/mL). Biflavanones such as rhusflavanone (9) and succedaneaflavanone (11), comprised of two naringenin units linked through either I-6-II-8 or I-6-II-6 linkages, were completely inactive.

[0134] Other structural characteristics were related to activity in our study. Methylation of the hydroxyl groups of the biflavanoids resulted in diminished activity. For instance, morelloflavone heptamethyl ether (8), volkensiflavone hexamethyl ether (6), and GB-1a hexamethyl ether (14), were inactive; all had exhibited moderate activity before alkylation. The fact that GB-1a-7″-O-glucoside (15), demonstrated no activity indicated that the 7″-hydroxyl group was especially important for anti-HIV-1 RT activity.

[0135] Six biflavanoids that were determined to be active in the HIV-1 RT enzyme assay were tested in human PBM cells infected with HIV-1 (strain LAV). These results are presented in Table 10. It has been observed that, although robustaflavone (3) exhibited significant inhibitory activity in the HIV-1 RT enzyme assay, it was found to be inactive in the assay for the PBM cells infected with HIV-1. However morelloflavone (7), in the whole cell assay, exhibited potent inhibitory activity with an EC50 (50% effective dose) value of 5.7 (8.0) &mgr;g/mL. Morelloflavone only possessed moderate activity in the anti-HIV-1 RT assay (IC50 64.7 &mgr;g/mL; 116.3 &mgr;M). This may suggest that the activity of these biflavanoids may be dependent upon different cellular mechanisms.

[0136] Other active compounds were hinokiflavone (4) and GB-1a (13), which exhibited good activity inhibiting viral replication in human PBM cells, but also high toxicity against uninfected PHA-stimulated human PBM cells. The other compounds (amentoflavone (1) and agathisflavone (2)) assayed in PBM cells appeared to either lack antiviral potency or display poor selectivity. From these results, it was concluded that biflavanoids comprised of flavanone (naringenin) and flavone (luteolin) via a I-3-II-8 bond demonstrate the most promising anti-HIV-1 activity.

[0137] In the past, some monoflavonoids have been reported to demonstrate anti-HIV activity. Baicalein (5,6,7-trihydroxyflavone), tiliroside (kaempferol 3-beta-D (6″-p-coumaroyl)glucoside), quercetin (3,3′,4′,5,7-pentahydroxyflavone), kaempferol (3,4′,5,7-tetrahydroxyflavone), and quercetagetin (3,3′,4′,5,6,7-hexahydroxyflavone) exhibited inhibitory activity against HIV-1 reverse transcriptase, whereas luteolin (3′,4′,5,7-tetrahydroxyflavone) and apigenin (4′,5,7-trihydroxyflavone) showed moderate to slight inhibition, and naringenin (4′,5,7-trihydroxyflavanone) was completely inactive.63,64,70 This revealed that the presence of both the unsaturated double bond between positions 2 and 3 of the flavonoid pyrone ring (e.g. flavone), and either the 3 hydroxyl groups introduced at the 5, 6, and 7 positions (bicalein) or the 3, 3′, and 4′ positions (quercetin) were a prerequisite for inhibition of RT activity.

[0138] In our study, apigenin exhibited moderate activity and naringenin demonstrated slight inhibition. Biflavanoids which consisted of two apigenin units (amentoflavone (1), agathisflavone (2), robustaflavone (3), and hinokiflavone (4)) demonstrated significant activity. Biflavanoids constructed of flavanone and flavone units (morelloflavone (7)) and biflavanone, linked through I-3-II-8 (GB-1a (13) and GB-2a (16)) were moderately active, and biflavanones linked through ring A of two naringenin units (rhusflavanone (9) and succedaneaflavanone (11)) were inactive. This structure-activity comparison again demonstrates that hydroxyl groups and at least one flavone unit in the biflavanoids are required for activity. A I-3-II-8 linkage is also necessary for biflavanones to exhibit activity. A firther conclusion is that previously active compounds become inactive when hydroxy groups are methylated.

EXAMPLE 10 Anti-Herpes Viral Activity of Biflavanoids

[0139] Anti-Herpes Viral Assay: The viruses used in the primary screen for anti-viral activity against herpes viruses consisted of: Herpes Virus 1 (HSV-1 E-377 strain), Herpes Virus 2 (HSV-2 MS strain), Cytomegalovirus (HCMV AD 169 strain), Varicella Zoster Virus (VZV Ellen Strain), and Epstein-Barr Virus (EBV), superinfection of Raji or Daudi cells with P3HR-1.

[0140] The assay for the inhibition of the cytopathic effect (CPE) for HSV, HCMV and VZV was as follows: Low passage human foreskin fibroblast cells were seeded in 96-well tissue culture plates 24 h prior to use, at a cell concentration of 2.5×104 cells/mL in 0.1 L of minimal essential medium (MEM) supplemented with 10% fetal bovine serum (FBS). The cells were then incubated for 24 h at 37° C. in a CO2 incubator. After incubation, the medium was removed and 100 mL of MEM containing 2% FBS was added to all but the first row. In the first row, 125 mL of the test compound was added in triplicate wells. Medium alone was added to both cell and virus control wells. The test compound in the first row was diluted serially 1:5 throughout the remaining wells by transferring 25 mL using a Cetus Liquid Handling Machine. After dilution of the compound, 100 mL of the appropriate virus concentration was added to each well, excluding cell control wells which received 100 mL of MEM. For HSV-1 and HSV-2 assays, the virus concentration utilized was 1000 PFUs per well. For CMV and VZV assays, the virus concentration added was 2500 PFUs per well. The plates were then incubated at 37° C. in a CO2 incubator for three days for HSV-1 and HSV-2, 10 days for VZV, or 14 days for CMV. After the incubation period, the media was aspirated and the cells stained with a 0.1% crystal violet solution for 30 min. The stain was then removed and the plates rinsed using tap water until all the excess stain was removed. The plates were allowed to dry for 24 h and then read on a Skatron Plate reader at 620 nm.

[0141] VZV Plaque Reduction Assay. Two days prior to use, HFF cells were plated into six-well plates and incubated at 37° C., with 5% CO2 atmosphere and 90% humidity. On the date of assay, the test compound was made up at twice the desired concentration in 2X MEM using six concentrations of the compound. The initial starting concentrations were usually from 200 ug/mL to 0.06 ug/mL. The VZV was diluted in 2X MEM containing 10% FBS to a desired concentration which would give 20-30 plaques per well. The media was then aspirated from the wells and 0.2 mL of the virus was added to each well in duplicate, with 0.2 mL of media being added to the drug toxicity wells. The plates were then incubated for 1 h with shaking every 15 min. After the incubation period, mean equal amount of 1% agarose was added to an equal volume of each test compound dilution. This provided final test compound concentrations beginning with 100 ug/mL and ending with 0.03 ug/mL, and a final agarose overlay concentration of 0.5%. The test compound agarose mixture was applied to each well in 2 mL volumes. The plates were then incubated, the stain aspirated, and plaques counted using a stereomicroscope at 10x magnification for ten days, after which the cells were stained with a 1.5% solution of neutral red dye. On days three and six an additional 1 mL overlay with equal amounts of 2X MEM and 1% agarose were added. At the end of the 4-6 h incubation period, the stain was aspirated and plaques counted using a stereomicroscope at 10x magnification.

Herpes viruses (HSV-1, HSV-2, HCMV, VZV, and EBV)

[0142] The results of the anti-herpes viruses activity assays of these biflavanoids are presented in Table 11. Among the compounds studied, only robustaflavone (3) exhibited significant inhibitory activities against HSV-1 and HSV-2 viruses. Activity values are measured by effective concentration (EC50) and cytotoxicity concentration (CC50) at which 50% of cells are free from pathogens or 50% of cells die. The values for robustaflavone (3) are an EC50 of 8.6 &mgr;g/mL and CC50>100 &mgr;g/mL, which results in a selectivity index of >11.6. The anti-viral activity of robustaflavone (3) against HSV-2 produced an EC50 value of 8.5 &mgr;g/mL, a CC50 of >100 &mgr;g/mL, and a SI of >11.8. Other results include amentoflavone (1) which demonstrated only slight activity against HSV-1. Volkensiflavone (5) exhibited weak inhibitory activity against both HCMV and VZV. Methylation of volkensiflavone (5) into volkensiflavone hexamethyl ether (6), resulted in the loss of activity, and a decrease in toxicity against HCMV, but an increase in activity and toxicity against VZV. Acetylation of rhusflavanone (9) to rhusflavanone hexaacetate (10) increased the activity and toxicity against HSV-1 and HSV-2. Acetylation of succedaneaflavanone (11) into succedaneaflavanone hexaacetate (12) led to a slight decrease of both activity and toxicity, and resulted in almost equal SI values. When assayed for activity against VZV, the acetylation product (12) resulted in an SI value which increased from <3 to 9.6. 11 TABLE 11 HSV-1 (HFF Cells) HSV-2 (HFF Cells) HCMV (HFF Cells) VZV (HFF Cells) CPE Inhibition CPE Inhibition CPE Inhibition Plaque Reduction EC50*1 CC50*2 EC50*1 CC50*2 EC50*1 CC50*2 EC50*1 CC50*2 Sample ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ug/ml ug/ml SI*3 ACT* 1.5 — 0.9 — — — — — 0.5 — — GVC* — >100 — — — 0.4 — — — — — Amentoflavone (1) 17.9 >100 >5.6 48.0 >100 >2.1 50.8 >100 >1.9 >4.0 9.3 >2.3 Agathisflavone (2) >100 >100 0 >100 >100 0 94.8 >100 >1.0 >4 12.0 <3.0 Robustaflavone (3) 8.6 >100 >11.6 8.5 >100 >11.8 54.8 >100 >1.8 — — — Hinokiflavone (4) >20 77.7 <3.9 >20 77.5 <3/9 >0.8 2.6 <3.2 >4.0 16.8 <4.2 Volkensiflavone (5) >100 >100 0 87.9 >100 >1.1 >4.0 16.8 <4.2 >20 80.0 <4.0 Volkensiflavone >100 >100 0 >100 >100 0 >100 >100 0 3.3 11.1 3.4 hexamethyl ether (6) Rhusflavanone (9) >100 >100 0 15.7 >100 >6.4 >4.0 13.7 <3.4 >4 16.0 <4 Rhusflavanone >4.0 18.5 <4.6 >4.0 18.5 <4.6 >4.0 13.4 <3.3 11.3 46.7 4.1 hexaacetate (10) Succedaneaflavone (11) >20 60.7 <3.0 >20 60.7 <3.0 >20 55.5 <2.7 >20 60.0 <3.0 Succedaneaflavone >4 17.7 <4.4 >4.0 17.7 <4.4 >4.0 14.4 <3.5 7.1 68.0 9.6 hexaacetate (12) *Positive control drug; *150% effective dose; *250% cell inhibitory (cytotoxic) concentration; *3selective index: CC50/EC50

EXAMPLE 11 In Vivo Evaluation of Robustaflavone in a Murine Influenza Model

[0143] In this Example, a series of in vivo experiments were run to determine if robustaflavone is efficacious against an experimentally induced influenza virus infection in mice. Prior to beginning this study, a series of preliminary experiments were run to determine the maximum tolerated dose of this compound in mice. Since the compound is not soluble in aqueous medium, it was suspended in 0.4% carboxymethylcellulose (CMC), a vehicle commonly used for water-insoluble compounds. When it was found that the compound was well tolerated at high dosages in this suspension, the question arose as to whether it was being adequately absorbed by the animal. Some studies were thus conducted using other vehicles in which the compound was more soluble. These vehicles included dimethylsulfoxide (DMSO), dimethyl formamide (DMF), and polyethylene glycol (PEG).

Materials and Methods

[0144] Animals: Female 13-15 g specific pathogen-free BALB/c mice were obtained form Simonsen Laboratories (Gilroy, Calif.). They were quarantined 24 h prior to use, and maintained on Wayne Lab Blox and tap water. After being infected, their drinking water contained 0.006% oxytetracycline (Pfizer, New York, N.Y.) to control possible secondary bacterial infections.

[0145] Virus: A/NWS/33 (H1N1) was obtained from K. W. Cochran, Univ. of Michigan (Ann Arbor, Mich.). A virus pool was prepared in MDCK cells; this was titrated in mice, ampuled, and stored at −80° C. until used.

[0146] Compounds: Robustaflavone was stored at room temperature until used. Ribavirin, used as a positive control, was obtained from ICN Pharmaceuticals (Costa Mesa, Calif.). Vehicles considered included DMSO (Sigma Chemical Co., St. Louis, Mo.), DMF (Sigma), PEG M.W. 200 (Aldrich Chemical Co. Milwaukee, Wis.), 0.4% CMC (Sigma) and 1-methyl-2-pyrrolidinone (MPD, Aldrich).

[0147] Arterial Oxygen Saturation (SaO2) Determinations: SaO2 was determined using the Ohmeda Blox 3740 pulse oximeter (Ohmeda, Louisville, Ohio)). The ear probe attachment was used, the probe placed on the thigh of the animal, with the slow instrument mode selected. Readings were made after a 30 second stabilization time on each animal. Use of this device for measuring effects of influenza virus on arterial oxygen saturation have been described.72

[0148] Lung Virus Determinations: Each mouse lung was homogenized and varying dilutions assayed in triplicate for infectious virus in MDCK cells as described previously.73

Experiment Design

[0149] 1. Toxicity Determination of robustaflavone in CMC Vehicle: The compound was suspended in 0.4% CMC at a concentration of 37.5 mg/mL to make a dosage of 500 mg/kg/day. It was injected i.p. into 2 mice daily for 5 days. The mice were weighed and deaths noted daily.

[0150] 2. Toxicity Determination of robustaflavone in 100% DMSO: The compound was dissolved in DMSO at a concentration of 25 mg/mL and in a later experiment in a concentration of 11.25 mg/mL to make dosages of 250 and 75 mg/kg/day, respectively. The higher dosage was injected i.p. into mice twice daily for 5 days in a volume of 0.1 mL/injection daily for 5 days in a volume of 0.05 mL/injection. As controls, mice were treated by the same treatment schedule with DMSO only in volumes of 0.1 or 0.05 mL/injection. Weight gain and mortality was determined in these animals.

[0151] 3. 3. Toxicity Determination of DMF and PEG only: DMF and PEG 200 in a concentration of 100% were injected i.p. into separate groups of mice daily for 5 days using a volume of 0.05 mL/injection. Again, effects on host weight and deaths of mice were monitored.

[0152] 4. Effect of robustaflavone in CMC or in DMSO on influenza virus infection in mice. In the study with CMC, robustaflavone was used in dosages of 200 and 100 mg/kg/day; using DMSO vehicle, the dosages were 75 and 37.5 mg/kg/day, with the compound administered i.p. twice daily for 5 days beginning 4 h pre-virus exposure. The mice were used in each dose to monitor effects on SaO2 and death; from an additional group of similarly infected and treated mice, 3 animals were killed on days 3, 5, 7 and 9 to assay for lung score (0=normal, 4=maximal consolidation), weight, and virus titer. Three to four mice were used as toxicity controls, which were weighed prior to treatment and again 18 h after treatment termination, and deaths noted daily. Ribavirin, dissolved in saline, was used in a dose of 75 mg/kg/day with the same treatment schedule. Three sets of virus controls were used: Infected-untreated, infected-treated with CMC only, and infected-treated with DMSO only. Twenty animals were used in each of these control groups to monitor SaO2 and death, with 3 additional mice taken in parallel with treated animals to determine effects on lung consolidation and virus titer. Two sets of normal controls were used; one group of three mice was weighed and held in parallel with the toxicity controls. From the second group three mice were killed on days 3 and 9 for comparison of lung score and weight.

[0153] Statistical Evaluation: Increase in survivor number was evaluated using chi square analysis with Yates' correction. Mean survival time increases, virus titer and SaO2 value differences were analyzed by t-test. Lung consolidation scores were evaluated by ranked sum analysis.

Results and Discussion

[0154] Toxicological Effects on Various Vehicles: The results of the various experiments with the vehicles considered are summarized in Table 12. CMC was the most well tolerated, followed by DMSO. DMF and PEG 200 were lethally toxic to the mice. One mouse died immediately following the day 4 i.p. treatment with DMSO; since this animal died instantly it is probable the death was due to penetration of an organ by the needle as it was administered into the peritoneal cavity. Using the 0.05 mL volume of DMSO, the animals appeared to tolerate this vehicle better than at 0.1 mL. DMF was highly lethal, killing both animals after two injections, and PEG 200 was only slightly better, with all mice dying after 3 injections.

[0155] Based on the above data, both CMC and DMSO were used as solvents for robustaflavone, the latter used in injection volumes of 0.05 mL.

[0156] Dose Range-Finding Studies with Robustaflavone in Mice: Using CMC as vehicle, robustaflavone appeared to be quite insoluble, with dense yellow particulate material seen in the formulation. When injected i.p. twice daily for 5 days, a dose of 200 mg/kg/day appeared reasonably well tolerated, the treated animals surviving therapy but losing 0.1 g of weight in the 5-day treatment period. The material was very soluble in DMSO, forming a clear solution. A 250 mg/kg/day dose injected i.p. twice daily for 5 days was lethally toxic to the mice, all animals dying by day 5 of treatment and a 6 g weight loss seen. The injection volume in this experiment was 0.1 mL, when the experiment was repeated using 0.05 mL injection volume, the dosage was lowered to 75 mg/kg/day. At this dose, all mice survived, although they lost 2 g of weight during the 5-day treatment period.

[0157] The data using CMC as vehicle suggests the compound was not being well absorbed in the animal, so for the antiviral experiment it was decided to use doses of 200 and 100 mg/kg/day. The DMSO studies indicated 75 mg/kg/day may be approaching the maximum tolerated dose, so that dose and 37.5 mg/kg/day were chosen for the in vivo antiviral experiment.

[0158] Effect of robustaflavone in DMSO on Influenza A Virus Infections in Mice: The results of this experiment are summarized in Table 14 and FIG. 1 through 4. It was found that the 75 mg/kg/day dose in this antiviral experiment was lethally toxic to the mice; the 37.5 mg/kg/day dose killed 2 of 3 toxicity control mice as well. Due to this apparent toxicity, the effects on survivors and SaO2 values were inconclusive. This excess toxicity did not correlate with the earlier-run range-finding study, although in the latter study marked weight loss was seen suggesting the compound was approaching a lethally toxic dose.

[0159] A review of FIG. 2 and 3, showing effects of treatment on lung scores and lung weights, indicates a significant effect of this compound on lowering lung scores and weights. This effect was dose-responsive, and suggests robustaflavone may have a significant influenza-inhibitory effect which may also be seen at a dose more well tolerated to the mice.

[0160] DMSO used alone was not lethal to the mice, but infected animals treated with DMSO only died approximately 2 days sooner than untreated infected controls (Table 12). This suggests the DMSO injection may result in an enhancement of the infection.

[0161] Ribavirin, run in parallel as a positive control, was highly active in inhibiting the infection using all evaluation parameters.

[0162] Effect of robustaflavone in CMC on influenza A virus infections in mice: The results of this study are seen in Table 14 and in FIGS. 5 through 8. Robustaflavone appeared to be well tolerated in this experiment, with all toxicity controls surviving and host weight gain approaching that seen with normal controls run in parallel observed. The therapy did not prevent death, but did increase mean survival times in a dose-responsive manner. SaO2 levels remained high in these treated animals as well (Table 14, FIG. 5).

[0163] Treatment with this compound also inhibited lung consolidation in a dose-responsive fashion as seen in FIGS. 6 and 7.

[0164] These data indicate that: 1) Robustaflavone can be inhibitory to the in vivo influenza infection and 2) there is apparently at least a partial absorption of the compound since the dose-responsive effects were seen.

[0165] It may be pertinent to note that two flavones have previously been reported to have influenza virus-inhibitory effects. 5,7,8,4′-Tetrahydroxyflavone was reported in 199274 to prevent viral proliferation in lungs of infected mice when the compound was administered either by the intranasal or oral routes. The related 8-methylether compound, 5,7,4′-trihydroxy-8-methoxyflavone, was similarly effective when administered intranasally or by the i.p. routes.74-77 Research by these investigators indicated the compounds reduce viral replication by inhibiting fusion of the virus with endosome/lysome membrane which occurs at an early stage of the virus infection cycle and may also inhibit budding of the progeny virus from the cells surface.77,78 The vehicle for these flavones was Na2CO3/saline.

Conclusion

[0166] Robustaflavone was evaluated against influenza A/NWS/33 (H1N1) virus infections in mice using two vehicles, 0.4% carboxymethylcellulose (CMC) and 100% dimethylsulfoxide (DMSO) Treatment was i.p. twice daily for 5 days beginning 4 h pre-virus exposure. The compound in DMSO was toxic to the mice at the two dosages employed, 75 and 37.5 mg/kg/day; despite this toxicity, significant reduction in lung consolidation was seen. When used in CMC, the doses of 200 and 100 mg/kg/day used were well tolerated and both inhibited lung consolidation and slowed the mean day to death of the animals. 12 TABLE 12 Toxicological Effects of CMC, DMSO, DMSF, and PEG 200 in BALB/c Mice1 Volume/ Mean Host Injection Surv/ Mean Day Wt. Vehicle (ml) Total To Death Change (g)2 0.4% 0.1 3 3 >21 1.7 Carboxymethyl- cellulose (CMC) 100% DMSO 0.1 2 2 >21 −.26 100% DMSO 0.05 1 2 4.0 −0.3 100 MF 0.05 0 2 1.0 7 100% PEC 200 0.08 1 2 5.0 −2.2 100% PEC 200 0.1 0 2 2.0 −2.8 1Treatment i.p. bif × 5. 2Maximum difference between initial weight and weight after treatment.

[0167] 13 TABLE 13 Effect of i.p. Treatment with Robustaflavone in DMSO Vehicle on influenza A (H1N1) Virus Infections in Mice Animals: 13-15 g female BALB/c Mice Treatment Schedule: bid × 5 beg −4 Virus: Influenza A (A/NWS/33 (H1N1), h pre-virus exposure i.n. Treatment route: i.p. Drug Diluent: Robustaflavone 0.4% Experiment Duration: 21 days DMSO; Ribavirin Saline Toxicity Controls Infected, Treated Dosage Surv/ Mean Weight Surv/ MSTb Mean Compound (mg/kg/day) Total Change (g)a Total (days) SaO2c (%) Robustaflavone 75 0/3 −1.7 0/9  3.2 70.6 37.5 1/3 −0.8 0/10 8.0 73.8 Ribavirin 75 3/3 −0.5  10/10** >21.0** 87.1** DMSO — — — 0/20 9.6 82.6 Untreated — — — 0/20 11.4 84.2 Normals — 3/3  2.0 — — 87.9 aDifference between initial weight at start of treatment and weight 18 h following final treatment of toxicity controls. bMean survival time of mice dying on or before day 21. cMean of days 3-10. **P < 0.01 compared to DMSO-treated controls.

[0168] 14 TABLE 14 Effect of i.p. Treatment with Robustaflavone in CMC Vehicle on influenza A (H1N1) Virus Infections in Mice Animals: 13-15 g female BALB/c Mice Treatment Schedule: bid × 5 beg Virus: Influenza A (A/NWS/33 (H1N1), -4 h pre-virus exposure i.n. Treatment route: i.p. Drug Diluent: Robustaflavone 0.4% Experiment Duration: 21 days CMC; Ribavirin Saline Toxicity Infected, Controls Treated Mean Weight Com- Dosage Surv/ Change Surv MSTb Mean pound (mg/kg/day) Total (g)a Total (days) SaO2c (%) Robusta- 200  3/3 1.5  0/9 11.1 84.6** flavone 1005  4/4 1.9  0/10 9.8 85.1** Ribavirin 75 3/3 −0.5   10/10** >21.0** 87.1** CMC — — —  0/16 9.3 80.4 Untreated — — —  0/20 11.4  84.2 Normals — 3/3 2.0 — — 87.9 aDifference between initial weight at start of treatment and weight 18 h following final treatment of toxicity controls. bMean survival time of mice dying on or before day 21. cMean of days 3-10. **P < 0.01 compared to CMC-treated controls.

CONCLUSION

[0169] Robustaflavone, a naturally occurring biflavanoid isolated from the seed kernel extract of Rhus succedanea, was found to be a potent in vitro inhibitor of hepatitis B virus (HBV) replication in chronically infected human hepatoblastoma 2.2.15 cells, with an effective concentration (EC50) of 0.25 &mgr;M, and a therapeutic index (IC50C50) of 153. These values were compared with penciclovir and lamivudine (3TC), which exhibited EC50 vlaues of 0.19 and 0.038 &mgr;M, respectively, and therapeutic indexes of 471 and 11200. Combinations of robustaflavone with penciclovir and lamivudine displayed synergistic anti-HBV activity, having the most pronounced effects when the combination ratios were similar to the ratio of EC50 potencies. Thus, a 1:1 combination of robustaflavone and penciclovir exhibited an EC50of 0.11 &mgr;M and a therapeutic index of 684, while a 10:1 combination of robustaflavone and limivudine exhibited an EC50 of 0.054 &mgr;M and a therapeutic index of 894. Measurement of extracellular and intracellular HBV DNA, HBV RNA and viral protein levels following exposure of 2.2.15 cells to robustaflavone indicated that only HBV DNA levels were affected, suggesting that inhibition of HBV DNA polymerase is the mechanism of action.

[0170] The results indicated that robustaflavone and robustaflavone tetrasulfate potassium salt were extremely effective anti-HBV agents. Robustaflavone also exhibited strong inhibitory effects against influenza A and influenza B viruses. Both hinokiflavone and robustaflavone demonstrated similar activity against HIV-1 RT, producing IC50 values of 35.2 &mgr;g/mL and 33.7 &mgr;g/mL, respectively. Amentoflavone, agathisflavone, morelloflavone, GB-1a and GB-2a were moderately active against HIV-1 RT, with IC50 values of 64.0 &mgr;g/mL, 53.8 &mgr;g/mL, 64.7 &mgr;g/mL, 127.8 &mgr;g/mL, and 94.6 &mgr;g/mL, respectively. Morelloflavone also demonstrated significant antiviral activity against HIV-1 (strain LAV in phytohemagglutinin (PHA)-stimulated human peripheral blood mononuclear (PBM) cells) at an EC50 value of 5.7 &mgr;M and an SI value (selectivity index) of approximately 10. The other biflavanoids were either slightly active or inactive against these viruses and HIV-1 RT.

[0171] Amentoflavanone (1), agathisflavone (2), volkensiflavanone (5), volkensiflavone hexamethyl ether (6), rhusflavanone (9), and succedaneaflavone (11) exhibited inhibitory activity against influenza B virus with the selective index (SI) of 178, 5.6, 34, ˜38, 9.3 and 15, respectively. Amentoflavone (1), and agathisflavone (2) also demonstrated anti-influenza A activity.

[0172] Robustaflavone (3) produced moderate inhibitory activity against both HSV-1 and HSV-2. Rhusflavanone (9) was active against HSV-2, while succedaneaflavanone hexaacetate (12) was moderately active against VZV.

[0173] Comparison of robustaflavone with a series of other naturally occurring biflavanoids and biflavanones, as well as several semi-synthetic derivatives, indicated that robustaflavone prossesses unique structural features that impart the observed antiviral activity. The sermi-synthetic derivatives robustaflavone hexa-O-acetate and robustaflavone hexa-O-methyl ether were approximately three-fold and 10-fold less potent with regard to anti-HBV activity, respectively, but neither of these derivatives exhibited cytotoxicity up to a concentration of 1000 uM. Volkensiflavone hexa-O-methyl ether, rhusflavanone hexa-O-acetate and succedaneaflavanone hexa-O-acetate were the only other non-robustaflavone analogues to inhibit HBV replication, but all possessed unacceptable selectivity indexes (1.3, 2.8 and 1.9, respectively). Interestingly, the parent compounds of the latter three hexa-O-acetate derivatives (volkensiflavone, rhusflavanone and succedaneaflavanone) were all inactive against HBV replication, in contrast to the relationship of robustaflavone with its hexaacetate.

References

[0174] 1. Hoofnagle, J. H. Chronic hepatitis B, N. Engl. J. Med. 1990, 323, 337-339.

[0175] 2. Aach, R. D. The treatment of chronic type B viral hepatitis. Ann. Intern. Med. 1988, 109, 88-91.

[0176] 3. Martin, P. and Friedman, L. S. In Innovations in Antiviral Development and the Detection of Virus Infections; T. M. Block; D. Junkind; R. L. Crowell; M. Dension; L. R. Walsh, Ed.; Plenum Press: New York, 1992, 111-120.

[0177] 4. Kaplan, M. M.; Webster, R. G. The epidemiology of influenza, Sci. Am., 1977, 236 (6), 88-105.

[0178] 5. Alexander, G. J.; Brahm, J.; Fagan, E. A.; Smith, H. M.; Daniels, H. M.; Eddleston, A. L.; Williams, R., Loss of HBSAg with interferon therapy in chronic hepatitis B virus infection. Lancet 1987, ii, 66-69.

[0179] 6. Hoofnagle, J. H.; Di Bisceglie, A. M. Antiviral therapy of viral hepatitis. In Antiviral Agents and Viral Diseases of Man; G. J. Galasso; R. J. Whiteley; T. C. Merigan, Ed; Raven Press: New York, 1972, 415-457.

[0180] 7. Yokosuka, O.; Omata, O. M.; Imazeki, F.; Okauda, K.; Summers, J. Changes of hepatitis B virus DNA in liver and serum caused by recombinant leukocyte interferon treatment: analysis of intrahepatic replicative hepatitis B virus DNA. Hepatology 1985, 5, 728-734.

[0181] 8. Doong, S. L.; Tsai, C. H.; Schinazi, R. F.; Liota, D. C.; Cheng, Y. C. Inhibition of the replication of hepatitis B virus in vitro by 2′,3′-dideoxy-3′-thiacytidine and related analogues. Pro. Natl. Acad. Sci. USA 1991, 88, 8495-99.

[0182] 9. Schlam, S. W.; de Man, R. A.; Heijtink, R. A.; Niesters, G. M. New nucleoside analogues for chronic hepatitis B. J. Hepatalogy 1995, 22, 52-56.

[0183] 10. van Leeuwen R.; Katlama, C.; Kitchen, V.; Boucher, C. A. B.; Tubiana, R.; McBride, M. Ingrand, D.; Weber, J.; Hill, A.; McDade, H.; Danner, S. A. Evaluation of safety and efficacy of 3TC (Lamivudine) in patients with asymptomatic or mildly symptomatic human immunodeficiency virus infection: A phase I/II study. J. Inf. Dis. 1995, 171, 1166-71.

[0184] 11. Hoffman, C. E. Amantadine HCl and related compounds. In Selective Inhibitors of Viral Functions; Carter, W. A., Ed.; CRC Press: Cleveland, 1973, 199.

[0185] 12. Dolin, R.; Reichman, R. C.; Madore, H. P.; Maynard, R.; Lindon, P. M.; Webber-Jones, J. A controlled trial of amantadine and rimandatine in the prophylaxis of influenza A infections. N. Engl. J. Med. 1982, 307, 580-584.

[0186] 13. Oxford, J. S.; Galbraith, A. Anti-influenza virus activity of amantadine: A selective review of laboratory and clinical data: In Viral Chemotherapy; Shugar D. Ed.; Pergamon Press, 1985, 169-254.

[0187] 14. Couch, R. B.; Jackson, G. G. Antiviral agents in influenza—Summary of influenza workshop VIII. J. Infect. Dis. 1976, 134,516-527.

[0188] 15. Bryson, Y. J.; Monahan, C.; Pollack, M.; Shields, W. D. A prospective double-blind study of side effects associated with the administration of amantadine for influenza A virus prophylaxis. J. Infect. Dis. 1980, 141, 543-547.

[0189] 16. Tsunoda, A.; Maasab, H. H.; Cochran, K. W.; Eveland, W. C. Antiviral activity of a-methyl-1-adamantane methylamine hydrochloride. In Antimicrob. Agents Chemother. 1966, 553.

[0190] 17. Tisdale, M.; Bauer, D. J. The relative potencies of anti-influenza compounds. Ann. N.Y. Acad. Sci. 1977, .284, 254-263.

[0191] 18. Degelau, J; Somani, S. K.; Cooper, S. L.; Guay, D. R. P.; Crossley, K. B. Amantadine-resistant influenza A in a nursing facility. Arch. Intern. Med. 1992, 152, 390-392.

[0192] 19. Hayden, F. G.; Belshe, R. B.; Clover, R. D.; Hay, A. J.; Oakers, M. G.; Soo, W. Emergence and apparent transmission of rimantadine-resistant influenza virus in families. N. Engl. J. Med. 1989, 321, 1696-1702.

[0193] 20. Mast, E. E.; Harmon, M. W.; Gravenstein, S.; Wu, S. P.; Arden, H. H.; Circo, R.; Tyszka, G.; Kendal, A. P.; Davis, J. P. Emergence and possible transmission of amantadine-resistant viruses during nursing home outbreaks of influenza A (H3N2). Am J. Epidemiol. 1992, 134, 988-997.

[0194] 21. Hayden, F. G.; Couch, R. B. Clinical and epidemiological importance of influenza A viruses resistant to amantadine and rimantadine. Rev. Med. Virol., 1992, 2, 89-96.

[0195] 22. Kimberlin, D. W.; Crampacker, C. S.; Straus, S. E.; Biron, K. K.; Drew, W. L.; Hayden, F. G.; McKinlay, M.; Richman, D. D.; Whitley, R. J. Antiviral resistance in clinical practice. Antiviral Res., 1995, 26, 423-438.

[0196] 23. Knight, V.; Gilbert, B. E. Ribavirin aerosol treatment of influenza. In Infectious Disease Clinics of North America, Vol 1.; Moellering, Jr.. Ed.; 1987, 441-57.

[0197] 24. Ray, C. G.; Icenogle, T. B.; Minnich, L. L; Copeland, J. G.; Grogan, T. M. The use of intravenous ribavirin to treat influenza virus-associated acute myocarditis. J. Infect Dis., 1989, 159, 829-836.

[0198] 25. Lin, Y. M.; Chen, F. C. Robustaflavone from the seed-kernels of Rhus succedanea. Phytochemistry, 1974, 13, 1617-1619.

[0199] 26. Mabry, T. J.; Markham, K. R.; Thomas, M. B. The Systematic Identification of Flavonoids; Springer, New York, 1970, 35-36.

[0200] 27. Qasim, M. A.; Roy, S. K.; Ilyas, M. Phenolic Constituents of Selaginellaceae. Indian Journal of Chemistry, 1985, 24B, 220.

[0201] 28. Arya, Ranjiana; Babu, Vikas; Ilyas, M.; Nasim, K. T. Phytochemical examination of the leaves of Anaeardium occidentale. J. Indian Chem. Soc., 1989, 66, 67-68.

[0202] 29. Xu, L.; Chem, Z.; sun, N. Zhiwu Xuebao, 1993, 35, 138-143.

[0203] 30. Lopez-Saez, J. A.; Perez-Alonso, M.; Negueruela, A. V. Biflavanoids of Selaginella denticulata growing in Spain. Naturforsch., 1994, 49c, 267-270.

[0204] 31. Silva, G. L.; Chai, H.; Gupta, M. P.; Farnsworth, N. R.; Cordell, G. A.; Pezzuto, J. M.; Beecher, C. W. W.; Kinghorn, A. D. Cytotoxic biflavanoids from Selaginella willdenowii. Phytochemistry, 1995, 40, 129-134.

[0205] 32. Sidwell, R. W.; Bailey, D. W.; Wong, M. H.; Huffman, J. H.: In vitro and in vivo sensitivity of a non-mouse-adapted influenza (Beijing) virus infection to amantadine and ribavirin. Chemotherapy, 1995: 41, 455-461.

[0206] 33. Anand, K. K.; Gupta, V. N.; Rangari, V.; Singh, B.; Chandan, B. K. Structure and hepatoprotective activity of a biflavonoid from Ganarium manil. Planta Medica, 1992, 58, 493-5.

[0207] 34. Baba, K.; Takeuchi, K.; Mitsunobu, D.; Maisugi, K. The structures of new biflavonoids from Daphne odora Thunb. Tennen Yuki Kagobutsu Toronkai Koen Yoshishu, 1987, 29, 668-75 (Japan).

[0208] 35. Bardos, T. J.; Schinazi, R. F. Ling, K.-H.; Heider, A. R. Structure-activity relationships and mode of action of 5-mercapto-substituted oligo- and polynucleotides as antitemplates inhibiting replication of human immunodeficiency virus type 1. Antimicrob. Agents Chemother., 1992, 36, 108-114.

[0209] 36. Barre-Sinoussi, F.; Chermann, J. C.; Rey, R.; Nugeyre, L. M. T.; Chamaret, S.; Gruest, J.; Dauguet, C.; Axler-Blin, C.; Vezinet-Brun, F.; Rouzioux, C.; Rozenbaum, W., and Montagnier, L. Isolation of a T-lymphotopic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science, 1983, 220, 868-871.

[0210] 37. Chen, F. C.; Lin, Y. M. Rhusflavanone, a new biflavanone from the seeds of wax tree. J. Chem. Soc., Perkin Trans., 1976, I, 98-101.

[0211] 38. Chen, F. C.; Lin, Y. M. Succedaneaflavanone—A new 6,6″-biflavanone from Rhus succedanea. Phytochemistry, 1975A, 14, 1644-1647.

[0212] 39. Chen, F. C.; Lin, Y. M.; Hung, J. G. Phenolic compounds from the heartwood of Gracinia multiflora. Phytochemistry, 1975B, 14, 300-303.

[0213] 40. Chen, F. C.; Lin, Y. M.; Hung, J. C. A new biflavanone glucoside from Garcinia multiflora. Phytochemistry, 1975C, 14, 818-820.

[0214] 41. Cholla, M. R.; Paya, M.; Alcaraz, M. J. Inhibitory effects of phenolic compounds on CCl4 induced microsomal lipid peroxidation. Experientia, 1991, 47, 195-199.

[0215] 42. Chou, T.-C.; Talalay, P. Quantitative analysis of dose-effect relationships: The combined effects of multiple drugs or enzyme inhibitors. Adv. Enz. Regul., 1984, 22, 27-35.

[0216] 43. Chou, J.; Chou, T.-C. Dose-effect analysis with microcomputers: Quantitation of ED50, LD50, synergism, antagonism, low-dose risk, receptor binding and enzyme kinetics. A computer software for Apple II series and IBM-PC and Instruction Manual. 1985, Elsevier Science Publishers, Cambridge, U.K.

[0217] 44. Furukawa, S. (1932) Sci. Papers Inst. Phy. Chem., 1932, 19, 27, (Japan).

[0218] 45. Furukawa, S. (1933) Sci. Papers Inst. Phy. Chem., 1933, 21, 273-278, (Japan).

[0219] 46. Gallo, R. C.; Salahuddin, S. Z.; Popovic, M.; Shearer, G. M.; Kaplan, M.; Haynes, B. F.; Palker, T. J.; Redfield, R.; Oleske, J.; Safai, B.; White, G.; Foster, P.; Markham, P. D. Frequent and detection and isolation of cytopathic retroviruses (HTLV-III) from patients with AIDS and at risk for AIDS. Science, 1984, 224, 500-503.

[0220] 47. Tan, G. T.; Pezzuto J. M.; Kinghorn, A. D. Evaluation of natural products as inhibitors of human immunodeficienct virus Type I (HIV-1) reverse transcriptase. J. Nat. Prod., 1991, 54, 143.

[0221] 48. Hayashi, T.; Morita, N. Mechanism of action of the antiherpesvirus biflavone ginkgetin. Antimicrob. Agents Chemother., 1992, 36, 1890-3.

[0222] 49. Iwu, M. M.; Igbokao, O. A.; Onwuchekwa, U. A; Okunii, C. O. Evaluation of the antihepatotoxic activity of the biflavonoids of Garcinia kola seed. J. Ethnopharmacol., 1987, 21, 127-38.

[0223] 50. Iwu, M. M.; Igbokao, D. A.; Okunji, D. D.; Tempesta, M. Antidiabetic and aldose reductase activities of biflavanones of Gracinia kola. J. Pharm. Pharmacol., 1990, 42, 290-2.

[0224] 51. Konoshima, T.; Takasaki, M.; Kozuka, M.; Lin, Y. M.; Chen, F. C.; Tokuda, H.; Matsumoto, T. Studies on inhibitors of skin tumors promotion (IV). Inhibitory effects of flavonoids on Epstein-Barr virus activation (1). Shoyakugaku Zasshi, 1988, 42, 343-6.

[0225] 52. Kozuka, M.; Tokuda, H.; Matsumoto, T. Hinokiflavone and kayaflavone as antiviral agents. Jpn. Kokai Tokkyo Koho JP 01,221,314 [89,221,314] (Cl. A61K31/35), Sep. 4, 1989, Appl. 88/44,251, Feb. 29, 1988.

[0226] 53. Lin, Y. M.; Chen. F. C.; Liang, C. M. Biflavonyls from drupes of Rhus succedanea. Phytochemistry, 1974A, 12, 276-7.

[0227] 54. Lin, Y. M.; Chen, F. C. Agathisflavone from the drupes of Rhus succedanea. Phytochemistry, 1974B, 13, 657-658.

[0228] 55. Lin, Y. M.; Chen, F. C. Robustaflavone from the seed kernels of Rhus succedanea. Phytochemistry, 1974C, 13, 1617-1619.

[0229] 56. Lin, Y. M.; Chen, F. C.; Lee, K. H. Hinokiflavone, a cytotoxic principle from Rhus succedanea and the cytoxicity of the related biflavonoids. Planta Medica, 1989, 55, 166-8.

[0230] 57. MAPE establishment (1987) Ger. Offen. DE 3,544,457 (Cl. C07D311/40), Jun. 19, 1987, Appl. Dec. 16, 1985).

[0231] 58. McDougal, J. S.; Cort, S. P.; Kennedy, M. S.; Cabridilla, C. D.; Feorino, P. M.; Francis, D. P.; Hicks, K.; Kalyanaramen, V. S.; Martin, L. S. Immunoassay for the detection and quantitation of infectious human retrovirus, lymphadenopathy-associated virus (LAV). J. Immun. Meth. 1985, 76, 171-183.

[0232] 59. Mora, A.; Paya, M.; Roips, K. Structure-activity relationships of polymethoxyflavones and other flavonoids as inhibitor of non-enzymatic lipid peroxidation. Biochem-Pharmacol., 1990, 40, 793-7.

[0233] 60. Murakami, A.; Ohigashi, H.; Jisaka, M.; Irie, R.; Koshimizu, K. Cancer Lett. (Shannon, Irel.), 1991, 58, 101-6.

[0234] 61. Nakazawa, K. The Chemical Structure of Ginkgetin. Gifu Yakka Diagaku. Kiyo, 1941, 12, 1, Chem. Abst. 59,2759d.

[0235] 62. Natarajan, S.; Murti, V. V. S.; Seshadri, T. R.; Ramaswamy, A. S. Some new pharmacological properties of flavonoids and biflavonoids. Current Science, 1970, 39, 553.

[0236] 63. Ono, K.; Nakane, H.; Jukushima, M.; Chgermann, J. K.; Barre-Sinlussi, F. Inhibition of reverse transcriptase activity by a flavonoid compound, 5,6,7-trihydroxyflavone. Biochem. Biophys. Res. Commu., 1989, 160, 982-7.

[0237] 64. Ono, K.; Nakane, H.; Jukushima, M.; Chgermann, J. K.; Barre-Sinlussi, F. Differential inhibitory effects of various flavonoids on the activities of reverse transcriptase and cellular DNA and RNA polymerases. Euro. J. Biochem., 1990, 190, 469-476.

[0238] 65. Okunji, C. O.; Iwu, M. M. Molluscicidal activitv of Garcinia kola biflavanone. Fitoterapia, 1991, 62, 74-6.

[0239] 66. Sanz, M. J.; Ferrandiz, M. J.; Cejudo, M.; Terencia, M. C.; Gil, B.; Bustos, G.; Ubeda, A.; Gunasegaran, R.; Alcaraz, M. M. Influence of a series of natural flavonoids on free radical generating systems and oxidative stress. J Xenobiotica, 1994, 24, 689-99.

[0240] 67. Schinazi, R. F.; Cannon, D. L.; Arnold, B. H.; Martino-Saltzman, D. Combination of isoprinosine and 3′-azido-3′-deoxythymidine in lymphocytes infected with human immunodeficiency virus type 1. Antimicrob. Agents Chemother., 1988, 32, 1784-1787.

[0241] 68. Schinazi, R. F.; Sommadossi, J. P.; Saalmann, V.; Cannan, M. W.; Hart, G.; Smith, G.; Hahn, E. Activity of 3′-azido-3′-deoxythimidine nucleotide dimmers in primary lymphocytes infected with human immunideficiency virus type 1. Antimicrob. Agents Chemother., 1990, 34, 1061-1067.

[0242] 69. Spira, T. J.; Bozeman, L. H.; Holman, R. C.; Warfield, K. T.; Phillips, S. K.; Feoprino, P. M. Micromethod for assaying the reverse transcriptase of LAV-HTIV-III/lymphadenopathy-associated virus. J. Clin. Microbiol., 1987, 25, 97-99.

[0243] 70. Tan, G. T.; Pezzuto, J. M.; Kinghom, A. D. Evaluation of natural products as inhibitors of human immunodeficiency virus type I (HIV-1) reverse transcriptase. J. Nat. Prod., 1991, 54, 143-154.

[0244] 71. Tokuda, F.; Matsumoto, T. Bilobetin as virus genome inactivating agent JPN. Kokai Tokkyo Koho JP 01 42,426 [89 42,426] (Cl. A61k31/35), Feb 14, 1989, Appl. 87/200,570, Aug. 11, 1987.

[0245] 72. Sidwell, R.; Huffman, R.; Gilbert. B.; Moscon, G.; Pedersen, R.; Burger, R.,; Warren, R. Utilization of pulse oximetry for the study of the inhibitory effects of antiviral agents on influenza virus in mice. Antimicrob. Ag. Chemother., 1992, 36, 473-476.

[0246] 73. Sidwell, R. W.; Huffman, J. H.; Call, E. W., Alaghamandan, H.; Cook, P. D.; Muce, R. K. Antiviral Res., 1995, 6, 343-353.

[0247] 74. Nagai, T.; Miyaichi, Y.; Tomimori, T.; Suzuki, Y.; Yamada, N. Inhibition of influenza virus sialidase and anti-influenza virus activit by plant flavonoids. Chem. Pharm. Bull., 1990, 38, 1329-32.

[0248] 75. Nagai, T.; Miyaichi, Y.; Tomimore, T.; Suzuki, Y.; Yamada, H. In vivo anti-influenza virus activity of plant flavonoids possessing inhibitory activity for influenza virus sialidase. Antiviral Res., 1990, 19: 207-17.

[0249] 76. Nagai, T.; Suzuki, Y.; Tomimore, T.; Yamada, H. Antiviral activity of plant flavonoid, 5,7,4′-trihydroxy-8-methoxyflavone, from roots of Scutellaria baicalenais against influenza A (H3N2) and B viruses. Biol. Pharm. Bull. 1995, 18, 295-9.

[0250] 77. Nagai, T.; Moriguchi, R.; Suzuki, Y.; Tomimori, T.; Yamada, H. Mode of action of the anti-influenza virus activity of plant flavonoid, 5,7,4′-trihydroxy-8-methoxyflavone, form the roots of Scutellaria baicalensis. Antiviral Res., 1995, 26, 11-25.

[0251] 78. Huang, L.; Kashiwade, Y.; Cosentino, L. M.; Fan, S.; Chen, C. H.; McPhail, A. T.; Fujoika, T.; Mihasha, K.; and Lee, K. H. Anti-AIDS Agents. 15. Synthesis and Anti-HIV Activity of Dihydroseselins and Related Analogs. J. Med. Chem., 1994,37, 3947-3955.

[0252] 79. Magri, N. J.; Kinston, D. G. I. Modified Toxols, 4. Synthesis and Biological Activity of Toxols Modified in the Side Chain. J Nat. Prod., 1987, 51, 298-306.

[0253] 80. Barron, D.; Ibrahim, R. K. Synthesis of flavanoid sulfates: 1. stepwise sulfation of position, 3,7, and 4′ using N,N′-dicyclohexycarbodiimide and tetrabutylamminium hydrogen sulfate. Tetrahedron, 1987, 43, 5197-5202.

[0254] 81. Korba, B. E.; Milman, G. A cell culture assay for compounds which inhibit hepatitis B virus replication. Antiviral Res., 1991, 15, 217-218.

[0255] 82. Korba, B. E.; Gerin, J. L. Use of a standardized cell culture assay to assess activities of nucleoside analogs against hepatitis B virus replication. Antiviral Res., 1992, 19, 55-70.

[0256] 83. Muller, C.; Bergmann, K. F.; Gerin, J. L.; Korba, B. E. Production of hepatitis B virus by stably transfected monocytic cell line U-937: a model for extrahepatic hepatitis B virus replication. J. Infect. Dis., 1992, 165, 929-933.

[0257] 84. Banhamou, Y.; Katlama, C.; Lunel, F.; Coutellier, A.; Dohin, E.; Hamm, N.; Tubiana, R.; Herson, S.; Poynard, T.; Opolon, P. Ann. Intern. Med., 1996, 125, 705-712.

[0258] 85. Korba, B. E.; Boyd, M. R. Antimicrob. Agents Chemother., 1996, 40, 1282-1284.

[0259] 86. Main, J.; Brown, J. L.; Karayiannis, P.; Georgiou, P.; Boyd, M.; Prince, W.; Thomas, H. J. Hepatol, 1994, 21 (Suppl 1.), S32.

[0260] 87. Ling, R.; Mutimer, D.; Ahmed, M.; Boxall, E. H.; Elias, E.; Dusheiko, G. M.; Harrison, T. J. Hepatology, 1996, 24, 711-713.

[0261] 88. Tipples, G. A.; Ma, M. M.; Fischer, K. P.; Bain, V.G.; Kneteman, N. M. Tyrrell, D. L. J. 1 Hepatology, 1996. 24, 714-717.

[0262] 89. Hammer, S. M.; Katzenstein, D. A.; Hughes, M. D.; Gundacker, H.; Schooley, R. T.; Haubrich, R. H.; Henry, W. K.; Lederman, M. M.; Phair, J. P.; Niu, M.; Hirsch, M. S.; Merigan, T. C. N. Engl. J. Med., 1996, 335, 1081-1090.

[0263] 90. Korba, B. E.; Gerin, J. L., Antiviral Res. 1992, 19, 55-70.

[0264] 91. Korba, B. E.; Milman, G., Antiviral Res., 1991, 15, 217-228.

[0265] 92. Korba, B. E., Antiviral Res. 1995, 29, 49-51.

[0266] 93. Korba, B. E.; Gerin, J. L., Antiviral Res. 1995, 28, 225-242.

[0267] 94. Seeger, C.; Summers, J.; Mason, W. S., Curr. Top. Microbiol. Immunol. 1991, 168, 41-60.

[0268] 95. Ganem, D., Curr. Top. Microbiol. Immunol., 1991, 168, 61-83.

[0269] 96. Belen'kii, M. S., Schinazi, R. F. “Multiple Drug Effect Analysis with Confidence Interval.” Antiviral Research 1994, 25, 1-11.

Claims

1. A method for treating or preventing a viral infection which comprises administering a composition comprising an antivirally effective amount of at least one biflavanoid and at least one antiviral agent.

2. The method according to claim 1, wherein said biflavanoid is selected from the group consisting of robustaflavone, hinokiflavone, amentoflavone, agathisflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, morelloflavone, GB-1a, GB-2a, derivatives or salts thereof.

3. The method according to claim 2, wherein said biflavanoid is robustaflavone.

4. The method according to claim 2, wherein said derivatives or salts thereof comprise alkyl ethers, esters, acid adducts, amines and sulfates.

5. The method according to claim 4, wherein said robustaflavone salt is robustaflavone tetrasulfate potassium salt.

6. The method according to claim 4, wherein said robustaflavone salt is robustaflavone hexa-O-methyl ether.

7. The method according to claim 4, wherein said robustaflavone salt is robustaflavone hexa-O-acetate.

8. The method according to claim 4, wherein said robustaflavone salt is robustaflavone tetrasodium salt.

9. The method according to claim 1, wherein said antiviral agent comprises AZT, ddC, ddI, D4T, lamivudine (3TC), alyclovir, gancyclovir, fluorinated nucleosides, TIBO derivatives, nevirapine, saquinavir, &agr;-interferon and recombinant CD4), imnmunostimulants (e.g., various interleukins and cytokines), immunomodulators and antibiotics (e.g., antibacterial, antifungal, anti-pneumocysitis agents).

10. The method according to claim 4, wherein said antiviral agent is 3TC.

11. The method according to claim 1, wherein said biflavanoid is robustaflavone and said antiviral agent is 3TC.

12. The method according to claim 1, wherein said viral infection is by an influenza virus.

13. The method according to claim 12, wherein said influenza virus is influenza A or influenza B virus.

14. The method according to claim 1, wherein said viral infection is by a hepatitis virus.

15. The method according to claim 14, wherein said hepatitis virus is hepatitis B virus.

16. The method according to claim 1, wherein said viral infection is by a Herpes virus.

17. The method according to claim 16, wherein said Herpes virus is HSV-1 or HSV-2 virus.

18. The method according to claim 1, wherein said viral infection is by a Varicella Zoster Virus (VZV).

19. The method according to claim 18, wherein said viral infection is by a respiratory virus.

20. The method according to claim 19, wherein said respiratory virus is a measles virus.

21. The method according to claim 1, wherein the viral infection is by a retrovirus.

22. The method according to claim 21, wherein the retrovirus is a human immunodeficiency virus (HIV).

23. The method according to claim 22, wherein human immunodeficiency virus (HIV) is HIV- 1.

24. A pharmaceutical composition for treating and/or preventing viral infections which comprises an antivirally effective amount of at least one substantially purified biflavanoid, at least one antiviral agent and a pharmaceutically acceptable carrier.

25. The composition according to claim 24 wherein said biflavanoid comprises robustaflavone, hinokiflavone, amentoflavone, agathisflavone, volkensiflavone, rhusflavanone, succedaneaflavanone, morelloflavone, GB-1a, GB-2a, derivatives or salts thereof.

26. The composition according to claim 25, wherein said biflavanoid is robustaflavone.

27. The composition according to claim 25, wherein said derivatives or salts thereof comprise alkyl ethers, esters, acid adducts, amines and sulfates.

28. The composition according to claim 27, wherein said derivatives or salts thereof is a robustaflavone salt.

29. The composition according to claim 25, wherein said robustaflavone salt is robustaflavone tetrasulfate potassium salt.

30. The composition according to claim 25, wherein said robustaflavone salt is robustaflavone hexa-O-methyl ether.

31. The composition according to claim 25, wherein said robustaflavone is robustaflavone hexa-O-acetate.

32. The composition according to claim 25, wherein said robustaflavone is robustaflavone tetrasodium salt.

33. The composition according to claim 24, wherein said antiviral agent comprises AZT, ddC, ddI, D4T, lamivudine (3TC), alyclovir, gancyclovir, fluorinated nucleosides, TIBO derivatives, nevirapine, saquinavir, &agr;-interferon and recombinant CD4), immunostimulants (e.g., various interleukins and cytokines), immunomodulators and antibiotics (e.g., antibacterial, antifungal, anti-pneumocysitis agents).

34. The composition according to claim 33, wherein said antiviral agent is 3TC.

35. The composition according to claim 24, wherein said biflavanoid is robustaflavone and said antiviral agent is 3TC.

36. A pharmaceutical composition for treating and/or preventing viral infections which comprises an antivirally effective amount of a biflavanoid derivative.

37. The composition of claim 36 wherein said biflavanoid derivative comprises robustaflavone hexa-O-methyl ether, robustaflavone hexa-O-acetate, and robustaflavone tetrasodium salt and a pharmaceutically acceptable carrier.

38. A method for treating or preventing a viral infection which comprises administering an antiviral effective amount of a biflavanoid derivative.

39. The method according to claim 38 wherein said biflavonoid derivative comprises robustaflavone hexa-O-methyl ether.

40. The method according to claim 38 wherein said biflavonoid derivative comprises robustaflavone hexa-O-acetate.

41. The method according to claim 38 wherein said biflavonoid derivative comprises robustaflavone tetrasodium salt.

42. A method for treating or preventing a hepatitis B viral infection which comprises administering a composition comprising an antivirally effective amount of robustaflavone hexa-O-acetate.

43. A method for treating or preventing a hepatitis B viral infection which comprises administering a composition comprising an antivirally effective amount of robustaflavone hexa-O-methyl ether.

44. A method for treating or preventing a hepatitis B viral infection which comprises administering a composition comprising an antivirally effective amount of robustaflavone tetrasodium salt.

45. The method according to claims 42, 43, or 44, further comprising administering at least one other antiviral agent.

46. The method according to claim 45, wherein said antiviral agent comprises AZT, ddC, ddI, D4T, lamivudine (3TC), alyclovir, gancyclovir, fluorinated nucleosides, TIBO derivatives, nevirapine, saquinavir, &agr;-interferon and recombinant CD4), immunostimulants (e.g., various interleukins and cytokines), immunomodulators and antibiotics (e.g., antibacterial, antifungal, anti-pneumocysitis agent).

47. The method according to claim 45, wherein said antiviral agent comprises AZT, ddC, ddI, D4T, lamivudine (3TC), alyclovir, gancyclovir, fluorinated nucleosides, TIBO derivatives, nevirapine, saquinavir, &agr;-interferon and recombinant CD4), immunostimulants (e.g., various interleukins and cytokines), immunomodulators and antibiotics (e.g., antibacterial, antifungal, anti-pneumocysitis agents).

48. The method according to claims 45, wherein said antiviral agent is 3TC.

49. A pharmaceutical composition for treating and/or preventing a hepatitis B viral infection which comprises an antivirally effective amount of robustaflavone hexa-O-acetate and a pharmaceutically acceptable carrier.

50. A pharmaceutical composition for treating and/or preventing a hepatitis B viral infection which comprises an antivirallv effective amount of robustaflavone hexa-O-methyl ether and a pharmaceutically acceptable carrier.

51. A pharmaceutical composition for treating and/or preventing a hepatitis B viral infection which comprises an antivirally effective amount of robustaflavone tetradsodium salt and a pharmaceutically acceptable carrier.

52. The pharmaceutical composition according to claims 49-51 further comprising at least one other anti-viral agent comprising AZT, ddC, ddI, D4T, lamivudine (3TC), alyclovir, gancyclovir, fluorinated nucleosides, TIBO derivatives, nevirapine, saquinavir, &agr;-interferon and recombinant CD4), immunostimulants (e.g., various interleukins and cytokines), immunomodulators and antibiotics (e.g., antibacterial, antifungal, anti-pneumocysitis agents).

Patent History
Publication number: 20020068757
Type: Application
Filed: Jan 17, 2001
Publication Date: Jun 6, 2002
Applicant: Advanced Life Sciences, Inc.
Inventors: Yuh-Meei Lin (Naperville, IL), David E. Zembower (La Grange Park, IL), Michael T. Flavin (Darien, IL), Ralph Schure (Darien, IL), Geng-Xian Zhao (Woodridge, IL)
Application Number: 09761909