Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATIONAL PARADIGM COMBATING HIV, HIV/HSV, OR HIV/HPV INFECTIONS IN HUMANS USING SMALL MOLECULAR WEIGHT COMPOUNDS FROM PLANTS
Document Type and Number:
WIPO Patent Application WO/2008/088806
Kind Code:
A1
Abstract:
Compounds, compositions and methods for the treatment of HIV/HSP/HPV, in particular, compositions and methods for a 3 part combination therapy for HIV/HSV/HPV, comprising a viral attachment inhibitor, a viral sequence integration inhibitor, and a proviral transcription inhibitor. The therapy is advantageous for the treatment of HIV infection, and is also effective for HSV and HPV infection. Also disclosed are novel viral attachment inhibitors and methods of use.

Inventors:
HUANG RU CHIH C (US)
ABD-ELAZEM IBRAHIM S (US)
Application Number:
PCT/US2008/000530
Publication Date:
July 24, 2008
Filing Date:
January 16, 2008
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV JOHNS HOPKINS (US)
HUANG RU CHIH C (US)
ABD-ELAZEM IBRAHIM S (US)
International Classes:
A61K31/00; C07H15/24
Foreign References:
US20030138394A12003-07-24
US20020151584A12002-10-17
US5837257A1998-11-17
US20040116411A12004-06-17
Attorney, Agent or Firm:
HOBBS, Ann S. (P.O. Box 34385Washington, District Of Columbia, US)
Download PDF:
Claims:

We Claim:

1. A pharmaceutical composition comprising a viral attachment inhibitor, a viral sequence integration inhibitor, and a proviral transcription inhibitor, in amounts effective to treat the symptoms of or inhibit the course of HIV infection, and a pharmaceutically acceptable carrier.

2. The pharmaceutical composition of claim 1 wherein the viral attachment inhibitor is a triterpenoid saponin or active derivative thereof.

3. The pharmaceutical composition of claim 2 wherein the triterpenoid saponin or derivative thereof is selected from the group consisting of arganine C (Rev. 1), Tieghemelin (Gen.l), acetylated arganine C or acetylated Tieghemelin.

4. The pharmaceutical composition of one of the preceding claims wherein the viral sequence integration inhibitor.is lithospermic acid (M 5 22) or lithospermic acid B (M 5 32).

5. The pharmaceutical composition of one of the preceding claims wherein the viral sequence integration inhibitor affects the enzymatic activity of all viral integrases

6. The pharmaceutical composition of one of the preceding claims wherein the proviral transcription inhibitor is a nontoxic water-soluble derivative of NDGA with proviral transcription inhibitor properties.

7. The pharmaceutical composition of one of the preceding claims wherein the proviral transcription inhibitor is G 4 N.

8. A kit comprising at least one unit dosage of a viral attachment inhibitor, a viral sequence integration inhibitor, and a proviral transcription inhibitor, in amounts effective to treat the symptoms of or inhibit the course of HIV infection.

9. The kit of claim 8 wherein the viral attachment inhibitor, viral sequence integration inhibitor, and proviral transcription inhibitor are in separate compositions.

10. The kit of claim 8 wherein the viral attachment inhibitor, viral sequence integration inhibitor, and proviral transcription inhibitor are in the same composition.

11. The kit of claim 8 wherein the viral attachment inhibitor is a triterpenoid saponin, an active derivative or component thereof.

12. The kit of claim 11 wherein wherein the triterpenoid saponin or derivative thereof is selected from the group consisting of arganine C (Rev. 1), Tieghemelin (Gen.l), acetylated arganine C or acetylated Tieghemelin.

13. The kit of one of the preceding claims wherein the viral sequence integration inhibitor, the viral integrase inhibitor, is lithospermic acid (Ms22) or lithospermic acid B (M 5 32).

14. The kit of one of the preceding claims wherein the inhibitor is a nontoxic water-soluble derivative of NDGA with proviral transcription inhibitor properties.

15. The kit of claim 14 wherein the inhibitor functions by interacting with promoter DNA selectively, said inhibitor causing the change of the chromatin conformation of the promoter so that it is unsuitable for proviral transcription.

16. The kit of one of the preceding claims wherein the proviral transcription inhibitor is G 4 N.

17. A method of treating HIV infection comprising the administration of effective amounts of a viral attachment inhibitor, a viral sequence integration inhibitor, and a proviral transcription inhibitor to an individual in need of treatment.

18. The method of claim 17 wherein the viral attachment inhibitor is a triterpenoid saponin, or an active derivative or component thereof.

19. The method of claim 18 wherein the triterpenoid saponin or derivative thereof is selected from the group consisting of arganine C (Rev. 1), Tieghemelin (Gen.l), acetylated arganine C or acetylated Tieghemelin.

20. The method of one of the preceding claims wherein the viral sequence integration inhibitor is lithospermic acid (M 5 22) or lithospermic acid B (M 5 32).

21. The method of one of the preceding claims wherein the proviral transcription inhibitor is a nontoxic water-soluble derivative of NDGA with proviral transcription inhibitor properties.

22. The method of one of the preceding claims wherein the proviral transcription inhibitor is

G 4 N.

23. The method of one of the preceding claims wherein administration is oral.

24. The method of one of the preceding claims wherein the effective amount is between 0.01 mg/kg/day and 100 mg/kg/day.

25. A triterpenoid saponin selected from the group consisting of acetylated arganine C or acetylated Tieghemelin.

26. A method of treating HIV infection comprising the administration of an effective amount of a triterpenoid saponin selected from the group consisting of arganine C (Rev. 1), Tieghemelin (Gen.l), acetylated arganine C or acetylated Tieghemelin to an individual in need of treatment.

27. A method of inhibiting HIV viral attachment comprising contacting a cell with a triterpenoid saponin selected from the group consisting of arganine C (Rev. 1), Tieghemelin (Gen.l), acetylated arganine C or acetylated Tieghemelin.

28. A pharmaceutical compound comprising the triterpenoid saponin of claim 25 and a pharmaceutically acceptable carrier.

Description:

COMBINATIONAL PARADIGM COMBATING VIRAL INFECTIONS IN HUMANS USING SMALL MOLECULAR WEIGHT COMPOUNDS

FROM PLANT

The research resulting in the invention described herein was supported in part by Grant No. NIH 1RO1DE12165 from the National Institutes of Health. The U.S. Government has certain rights in the invention.

BACKGROUND A recent WHO report has stated that there are 39.5 million people currently HFV positive worldwide. Of these, 4.3 million were infected in the year 2006 alone. AIDS deaths reached 2.9 million in 2006, which is the highest ever reported in any year. This number would have been even higher but for the recent progress made in providing the highly active antiretroviral therapy (HAART) to a large population of AIDS patients in underdeveloped countries. Nevertheless, there are still 40,000 new AIDS cases a year in the U.S. even with the HAART. Thus, alarmingly AIDS is on the rise worldwide (1).

Great effort has been focused on development of HFV-preventive vaccine during the past 20 years. However such vaccine research has met with unusual obstacles. As described in a recent article by Robert Gallo (2), it is not appropriate to develop HIV vaccines using attenuated live HIV because of the inherent danger of causing AIDS. Furthermore, there is no useful small animal model for studying HIV infection. The SIV/monkey model, while offering limited information on the mode of viral actions, is expensive in operation and generally not available to investigators. From many studies, we have also learned that the gpl20 based HIV vaccine was not overly effective, and does not protect all strains of viruses containing highly variable regions of the V 3 loops. In addition, all recombinant HIV strains occur in vivo at an exceedingly fast rate.

The anti HIV compounds that are currently being used to treat AIDS patients include several reverse transcriptase inhibitors: zidovudine, didanosine, zalcitabine, stavadine, lamivudine and nevirapapine; and protease inhibitors: saquinavir, ritonavir, indinavir and others. It has recently been observed that these drugs, while very potent suppressers of wild type viruses, gradually lose their effectiveness with the appearance of a group of populated viral mutants.

Thus, the development of alternative therapeutic methods that inhibit HIV infection and protect host cell mediated immunity (CMI) is urgently needed.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure IA. Structures of Rev.1, Gen 1, A-Rev and A-Gen

Figure IB. Antiviral Entry activity in HFV-based Cell-Cell Fusion Assay

Figure 2. Drug Effect on the Infectivity of HIV-I. A. HIV-I Attachment. B. HIV Infection.

Figure 3. Drug Effect on Replication of Infectious Live HIV-I Figure 4. Elimination of the Hemolytic Effect of Gen.1 by Acetylation of C28 Sugars

Figure 5. Inhibition of HIV-I Replication and Maintained the Viability of the Treated Cells with Acetylated Gen 1

Figure 6. Effect of Acetylated Gen.l on HIV-I and CD 4 + Cells. A. Infectivity Assay by X-GaI

Staining. B. Effect of Acetylated Gen.l on Live HIV-I replication and Infection. C. Effect of Acetylated Gen.1 on CD 4 + Cell Viability

Figure 7. Inhibition of HIV-I Replication by Non-Toxic Integrase Inhibitors M 5 22 and M 5 32

Figure 8. Effect of M 5 22 and M 5 32 on Five Strains of HIV-I

Figure 9. Ms22 Targets the Core Domain of HIV-I Integrase

Figure 10. Chemical structures of M 5 22 (a) and M 5 32 (b) Figure 11 A-D. Molecular Structures of MaU, M 4 N, G 4 N and P 4 N

Figure 12. Inhibition of Three Different Strains of HSV-I in Vero Cells

Figure 13. Inhibition of Acyclovir-resistent Strains of HSV-I by G 4 N and Acyclovir

Figure 14. Comparative Potency (IC 50 ) Between M 4 N and Acyclovir at Different HSV Passages of Drug Treatment Figure 15. Efficacy of G4N and P4N Against HSV-2 in a Mouse Vaginal Model

Figure 16. Control of HIV Infection by Blocking Three Steps of the Life Cycle

Figure 17. The Three Compounds for Combination Therapy of HIV-I

Figure 18. Inhibition of HIV-I Replication by Plant Compounds Either Alone or In Combination

DESCRIPTION

Described herein are compounds, compositions and methods for the treatment of HIV/HSP/HPV. In particular, disclosed are compositions and methods for a 3 part combination

therapy for HIV/HS V/HPV, comprising a viral attachment inhibitor, a viral sequence integration inhibitor, and a proviral transcription inhibitor. The therapy is particularly advantageous for the treatment of HIV infection, and is also effective for HSV and HPV infection that is often present in HIV patients. Also disclosed are novel viral attachment inhibitors and methods of use.

Thus, the pharmaceutical compositions described herein for treatment of HFV infection, in the presence or absence of HSV and/or HPV infection include a viral attachment inhibitor, e.g. a triterpenoid saponin, such as arganine C (Rev. 1), Tieghemelin (Gen.l), or acetylated Tieghemelin, a viral sequence integration inhibitor, such as lithospermic acid (M 5 22) or lithospermic acid B (M 5 32), and a proviral transcription inhibitor, such as G 4 N or other nontoxic water-soluble derivative of NDGA with proviral transcription inhibitor properties. It has been found that this combination of drugs can block HIV-I production in cultured CD 4 + cells synergistically against a wide variety of HIV strains. The combination is equally active against mutant HIV strains that are highly resistant to currently available drugs against HIV protease and reverse transcriptase as compared to wild-type virus. Although the inventors are not bound by any particular theory, it is believed that the compositions and methods of the invention act by attacking the viral growth cycle at three different stages, i.e. by preventing HIV attachment to cell membranes, preventing viral sequence integration, and preventing proviral transcription. Effective amounts of the pharmaceutical components may be administered together in a single pharmaceutical composition, or separately, in two or three pharmaceutical compositions, either simultaneously, or at different times.

The pharmaceutical compositions described above can also be used in combination with other known antiviral and anti-HIV/HSV/HPV pharmaceuticals. For example, the compounds can be interchanged with other known HFV compounds and agents in the same classes (i.e. viral attachment inhibitors, viral sequence integration inhibitors, and proviral transcription inhibitors) to obtain the disclosed combination pharmaceutical, or other HIV therapeutic compounds and agents can be used to supplement the combination therapy. Examples of such FDA approved compounds and agents include entry and fusion inhibitors, such as Enfuvirtide (Fuzeon); non- nucleoside reverse transcriptase inhibitors, such as Delavirdine (Rescriptor), Efavirenz (Sustiva), Nevirapine (Viramune); nucleoside reverse transcriptase inhibitors, such as Abacavir (Ziagen), Abacavir / Lamivudine / Zidovudine (Trizivir), Abacavir/Lamivudine (Epzicom), Didanosine (Videx, Videx EC), Emtricitabine (Emtriva), Emtricitabine/Tenofovir (Truvada) , Lamivudine

(Epivir), Lamivudine/Zidovudine (Combivir), Stavudine (Zerit), Tenofovir disoproxil fumarate (Viread), Zalcitabine (Hivid), Zidovudine (Retrovir); protease inhibitors, such as Amprenavir (Agenerase), Atazanavir (Reyataz), Darunavir (Prezista), Fosamprenavir (Lexiva), Indinavir (Crixivan); Lopinavir/Ritonavir (Kaletra); Nelfinavir (Viracept); Ritonavir (Norvir), Saquinavir (Fortovase, Invirase) and ipranavir (Aptivus). Examples of current experimental compounds and agents for HIV treatment include entry and fusion inhibitors, such as AMD070, BMS- 488043, Fozivudine tidoxil, GSK-873,140 (aplaviroc), PRO 140, PRO 542, Peptide T, SCH- D (vicriviroc), TNX-355, and UK-427,857 (maraviroc); integrase inhibitors, such as GS 9137, MK-0518, Microbicides, BMS-378806, C3 IG, Carbopol 974P (BufferGel), Carrageenan (Carraguard), Cellulose sulfate (Ushercell), Cyanovirin-N, Dextran sulfate, Hydroxyethyl cellulose, PRO 2000, SPL7013, Tenofovir, and UC-781; non-nucleoside reverse transcriptase inhibitors, such as Calanolide A, TMC 125 (etravirine); and nucleoside reverse transcriptase inhibitors such as AVX754 (apricitabine), Alovudine, Amdoxovir, Elvucitabine, KP- 1461, and Racivir.

Thus, the treatment methods encompass a method of treating HIV, HIWHS V or HIV/HPV in humans comprising administering an effective amount of the pharmaceutical composition(s) described above to inhibit the growth cycle of HIV, and/or treat HIV symptoms in a person in need of treatment. Symptoms which may be alleviated by the pharmaceutical compounds and compositions include a low CD 4 + count, sore throat, mouth sores, aching or stiff muscles, headache, diarrhea, swollen lymph glands, fever, fatigue, rash, including seborrheic dermatitis, frequent vaginal yeast infections.

Most, if not all, viral mutants, which include those explicatively active viral mutants, are host dependent in their replications. Host proteins, unlike viral proteins, when not under mutational pressure, are in general structurally invariable. Thus, compounds that block the usage of these cellular factors and membrane proteins at different stages of viral life cycle are likely to be good candidates as mutation-insensitive antiviral drugs. The work leading to the compositions and therapies described herein was initiated to specifically search for small chemical compounds that inhibit three cellular dependent steps of viral life cycle: to protect CD 4 + cells against viral entry, to prevent chromatin dependent viral integration and proviral transcription of both wild type and mutant strains of HIV by changing the chromatin structure of HIV LTR which serves as

promoter for such proviral transcription. This has resulted in a combinational drug design that protects host T lymphocytes against HIV with high efficacy and low drug resistance. Medicinal plants selected for have been based on three criteria: the crude materials of such plants must already be known as herbages which have been consumed by a large human population for thousands years, thus not likely to be overly toxic in long terms. Crude extracts must show significant antiviral activity at a concentration not damaging to the testing CD 4 + cells and such plant materials should be available in large quantities suitable for pure compound isolations at low cost. Using this approach, we isolated and chemically modified a series of compounds which show specific antiHIV activities by preventing HIV attachment to CD 4 + cells, by preventing HIV viral sequence integration, and by preventing HIV proviral transcription (3-6, 8- 11, 15-23, 25). We have found these compounds (e.g. Gen-1, M 5 22/M 5 32 and G 4 N respectively), when tested in combination, can block HIV-I production in culture CD 4 cells synergistically against a variety of HIV strains. They are equally active in targeting mutant strains that are highly resistant to currently available drugs against HIV protease and reverse transcriptase as compared to the wild type viruses. Furthermore, the combination is effective against other viral infections, such as HSV and HPV.

Viral attachment inhibitors are intended to include pharmaceutically acceptable compounds that inhibit the attachment of a virus (e.g. HIV, HSV, or HPV) to a cell surface.

Viral sequence integration inhibitors are intended to include any pharmaceutically acceptable compound that inhibits viral sequence integration in a human chromosome. Examples are lithospermic acid (M 5 22) and lithospermic acid B (M 5 32). These, and examples of other suitable compounds, are described in detail in USP 7,071,227 and more recently in Nunthaboot et al. European J. of Medicinal Chemistry, Article in Press 2006, available online @ www.sciencedirect.com.

Transcription inhibitors, for example G 4 N, work by blocking transcription factor SPl from binding to a variety of promoters of HIV, HSV and HPV genes. These drugs were found to be able to suppress the expression of viral growth related genes and growth of these three viruses (3). Isolation and chemical synthesis and modification of these plant-based compounds for

antiviral testing in animals are the next logical step prior to engaging in phase I clinical trials for HIV, HIV/HSV or HIV/HPV infected patients (3,4,5,10,15,21,25).

The invention is to be understood as not being limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.

Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.

All publications mentioned herein, including patents, patent applications, and journal articles are incorporated herein by reference in their entireties including the references cited therein, which are also incorporated herein by reference.

The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.

The compositions herein are formulated in accordance to the mode of potential administration. Thus, if the composition is intended to be administered intranasally or by inhalation, for example, the composition may be a converted to a powder or aerosol form, as conventional in the art, for such purposes. Other formulations, such as for oral or parenteral delivery, are also used as conventional in the art.

Compositions for administration herein may form solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders.

Definitions:

By "HIV" is meant Human Immunodeficiency Virus, including all strains of HIV-I and HIV-2 that infect humans.

By "HSV" is meant Herpes Simplex Virus, including all strains of HIV-I and HIV-2 that infect humans.

By "HPV" is meant all strains of Human papilloma virus.

"G 4 N" means meso-l,4-bis[3,4-(dimethylaminoacetoxy)phenyl]-(2R,3S)dimeth ylbutane, either as a free base or preferably as a pharmaceutically acceptable salt, such as the hydrochloride salt.

"NDGA" means nordihydroguaiaretic acid. In addition to G 4 N, suitable NDGA derivatives are described in U.S. Pat. Nos. 5,663,209, 6,214,874, 6,291,524, 6,365,787, 6,417,234, and 6,608,108.

By "derivative" is meant a compound with additions or substitutions, such as the addition of an acetyl group to a triterpenoid saponin, as disclosed herein. Other groups contemplated to be added are lower alkyl, acyl, alkoxy and the like (Ci -6 ). "Active derivatives" are those retaining the essential pharmaceutical properties of the parent compound (e.g. preventing HIV attachment to cell membranes, preventing viral sequence integration, or preventing proviral transcription). Derivatives may also consist of portions of the parent compound, wherein various substituents have been removed, or may consist of a particular substituent, if active (e.g. the substituent sugar, in the case of the triterpeoid saponins or by adding methyl groups to dihydroxyl-pheny . rings in the case of M 5 22 and M 5 32 or by attaching one to four piperiino groups in the case of making water soluble NDGA derivatives PiN, P 2 N, P 3 N and P 4 N).

Buffers suitable for use herein include any buffer conventional in the art, such as, for example, Tris, phosphate, imidazole, and bicarbonate.

As used herein, the terms "treatment," "treating," etc., refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a condition or disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a condition or disease and/or any adverse affect attributable to the condition or disease. "Treatment," thus, for example, covers: (a) preventing the condition or disease from occurring in an individual who is predisposed to the condition or disease but has not yet been diagnosed as having it; (b) inhibiting the condition or disease, such as, arresting its development; and (c) relieving, alleviating or ameliorating the condition or disease, such as, for example, causing regression of the condition or disease.

The term "pharmaceutically acceptable carrier" refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any conventional type. A "pharmaceutically acceptable carrier" is non-toxic to recipients at the dosages and concentrations employed, and is compatible with other ingredients of the formulation. For example, the carrier for a formulation containing the present therapeutic compounds and compositions preferably does not include oxidizing agents and other compounds that are known to be deleterious to such. Suitable carriers include, but are not limited to, water, dextrose, glycerol, saline, ethanol, buffer, dimethyl sulfoxide, Cremaphor EL, and combinations thereof. The carrier may contain additional agents such as wetting or emulsifying agents, or pH buffering agents. Other materials such as anti-oxidants, humectants, viscosity stabilizers, and similar agents may be added as necessary.

Pharmaceutically acceptable salts herein include the acid addition salts (e.g. formed with a free amino group) and which are formed with inorganic acids, including, but not limited to hydrochloric or phosphoric acids, or such organic acids as acetic, mandelic, oxalic, and tartaric. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, and histidine.

The term "pharmaceutically acceptable excipient," includes vehicles, adjuvants, or diluents or other auxiliary substances, such as those conventional in the art, which are readily available to the public. For example, pharmaceutically acceptable auxiliary substances include pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like.

As used herein, the singular forms "a", "an", and "the" include plural forms unless the context clearly dictates otherwise. Thus, for example, reference to "a compound" includes a plurality of such compounds.

Formulations, Dosages, and Routes of Administration

As mentioned above, effective amounts of the pharmaceutical compounds are administered to an individual, where "effective amount" means a dosage sufficient to produce a desired result. In some embodiments, the desired result is at least a stabilization or increase in CD4+ count in a patient.

Typically, the compositions of the instant invention will contain from less than about 1% up to about 99% of the active ingredient(s). The appropriate dose to be administered depends on the subject to be treated, such as the general health of the subject, the age of the subject, the state of the disease or condition, the weight of the subject, etc. Generally, between about 0.1 mg and about 500 mg or less may be administered to a child and between about 0.1 mg and about 5 grams or less may be administered to an adult. The pharmaceutical compounds can be administered in a single or, more typically, multiple doses. They may be formulated together into a single composition, or administered separately, either simultaneously or at different times. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. Other effective dosages can be readily determined by one of ordinary skill in the art through routine trials establishing dose response curves. The amount of compound to be administered will, of course, vary depending upon the particular compound. Typical dosages of the therapeutic compounds are expected to be between 0.01 and lOOmg mg/kg/day, more preferably between 0.1 and 10 mg/kg/day, or between 0.1 and 1 mg/kg/day. It will be clear to those of skill in the art that for the combination pharmaceutical composition and method of treatment the dosage for each compound can be optimized, and equal amounts need not be administered.

The frequency of administration of the therapeutic compounds and compositions, as with the doses, will be determined by the medical practitioner based on age, weight, disease status, health status and patient responsiveness. Thus, the compounds/compositions may be administered one or more times daily, weekly, monthly or as appropriate as conventionally determined. The therapeutic compounds and compositions may be administered intermittently, such as for a period of days, weeks or months, then not again until some time has passed, such as 3 or 6 months, and then administered again for a period of days, weeks, or months.

The therapeutic compounds of the present invention can be incorporated into a variety of formulations for therapeutic administration. More particularly, the compounds can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, aerosols, liposomes, nanoparticles, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.

As such, although administration of the therapeutic compounds may be achieved in various ways, such as oral, buccal, rectal, intranasal, intravenous, intra-arterial, intra-tracheal, intraventricular, intracranial, interstitial, transdermal, etc., or by inhalation or implantation, most preferably, the therapeutic compounds are administered orally or intravenously.

In pharmaceutical dosage forms, the compounds may be administered in the form of their pharmaceutically acceptable salts, or they may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.

For oral preparations, the therapeutic compounds can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as

talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.

The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are conventional in the art. Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle may contain minor amounts of auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents or emulsifying agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985. The composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the individual being treated.

The therapeutic compounds can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or non-aqueous solvent, such as vegetable or other similar oils, including corn oil, castor oil, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.

The therapeutic compounds can be utilized in aerosol formulation to be administered via inhalation, e.g. by formulation into pressurized acceptable propellants such as dichlorodifiuoromethane, propane, nitrogen and the like.

Furthermore, the therapeutic compounds can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. The compounds of the present invention can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.

Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or

suppository, contains a predetermined amount of the composition containing one or more inhibitors. Similarly, unit dosage forms for injection or intravenous administration may comprise the inhibitor(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.

The term "unit dosage form," as used herein, refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of compounds of the present invention calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for the novel unit dosage forms of the present invention depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the patient.

Kits with multiple or unit doses of the pharmaceutical compounds, are included in the present invention. Such kits, in addition to the containers containing the multiple or unit doses of the pharmaceutical compositions, optionally include an informational package insert with instructions describing the use and attendant benefits of the drugs in treating the diseases/conditions.

As will be evident to those of skill in the art, the compositions and methods described herein can also be used with pharmaceuticals used to treat opportunistic infections and the like which occur frequently in HIV patients.

This application claims priority to U.S. provisional application no. 60/880,388, filed January 16, 2007, which is hereby incorporated herein by reference.

EXAMPLES Example 1

Description of HIV Entry Inhibitors from Tieghemella heckeliϊ. Optimization of a new antiviral saponin, the acetylated tieghemella (O Aceto-Gen-1) against HIV-I viral entry and infection. a. Isolation and chemical characterization of two antiviral saponins from Tieghemella heckelii

Tieghemella heckelii (Sapotaceae, Th) is a large tree found in the West African rain forest. In January and February, the plant blossoms and develops ovoid fruits of 8 cm length and 7 cm diameter, which ripen in July-October. Many parts of the tree are used in traditional medicine. We have used a series of high-speed counter-current chromatography (HSCC), viral entry and infectivity assay and HIV-based cell-cell fusion assay to isolate specific HIV viral entry inhibitors from the ovoid fruits of the plants. The procedures for the isolation have been described in detail (4). Two pure saponin compounds, Arganine C (Rev 1) and Tieghemella (Gen. 1) have been isolated with the yield of 0.48% and 0.6% respectively. Rev.l the more active of the two has an IC 5O of 2.8μM while Gen-1 has an IC 50 of 17μM (Fig 1).

b. The C28 side chain sugar of Rev-1 and Gen-1 is essential for their antiviral activity. hi HIV infection, syncytia are the result of viral attachment and cell membrane fusion which create giant multi-nucleated cells consisting of both infected and uninfected cells. Rev. 1 and Gen. 1 have been shown to suppress syncytium formation resulting from co-culture of HeLa-CD4 + cells and a CHO-WT cell line expressing wild type HIV-I envelope (5). We found that arganine C (Rev-1) strongly inhibited HIV entry into CHO cells. Tieghemella (Gen-1) is less potent although less toxic than Rev-1. Rev-1 and Gen-1 are structurally identical except for one attached group at C23 (Fig IA). C23 glucose for Rev-1 and glucuronic acid for Gen-1 (4, 5). Their antiviral activity requires the C28 sugar chain. Removal of the four-sugar chains from Rev-1 and Gen-1 to give 16α-hydroxyprotobassic 3-0-β-D-glycopyransoide and 16α-hydroxyprotobassic acid 3-0-β-D- glucuronopyranoside respectively caused total loss of antiviral entry activity in both cases (A- REV and A-GEN). The four sugars in the substituent at the C28 position are L-arabinose, L- rhamnose, D-xylose and L-rhamnose (Fig IB).

c. Rev-1 and Gen-1 target HIV-I at the viral attachment step to the HeLa CD4 + .

To determine whether the two saponins act exclusively at the attachment step, target HeLa CD4 + cells were initially pre-cooled at 4 0 C before viral exposure to exclude the possibility of virus internalization. Each compound, at graded concentrations, was then mixed with HIV particles and added to target cells in cold media. After a 30-minute attachment period, the cells were profusely washed to remove unbound virus. The amount of viral capsid in cell lysates was measured and a dose-dependent inhibition of attachment by Rev.l and Gen.l was observed (Fig. 2A). The two compounds exhibited somewhat variable activities at low concentrations (<10μM)

which became indistinguishable at the maximum concentration (20μM). In contrast, when the side chain sugar of Gen- 1 at C-28 was cleaved, the resultant sapogenin (Agen) was totally devoid of activity. Similar results were obtained in an HIV infectivity assay when target HeLa CD4 + cells were challenged with virus particles and P24 antigen was monitored 48 hours later (Fig 2B). No significant difference was found in the activity as also shown by the cell-cell fusion assay (Fig 1). These inhibitory studies were repeated with infectious HIV virus tested under P 3 facilities. H9 cells (4 x 10 5 ) were first incubated with compound and then infected with virus for one hour at 37° C in CO 2 incubator to permit virus fusion and entry. The cells were then centrifuged and washed 3 times with phosphate buffered saline (PBS) to remove the unbound virus as described previously (6,7). The viral entry was determined by p24 antigen- capture assay. For the drug effect on HIV-I replication and infection, the culture was incubated for 8 days. Viral replication was determined by the amount of HIV-I p24 antigen in culture supernatant after 8 days of virus infection in H9 cells using the HIV/p24 monoclonal antibody assay (8). Fig. 3 describes the effect of Rev-1 and Gen-1 on viral replication and infection of the cells. Only low concentrations were used for testing to minimize the cellular toxicities of the drug.

d. Acetylation of C28 sugars of Gen-1 abolishes its hemolytic effect and increases its antiviral activities without cellular toxicity.

We have found that Rev-1 and Gen-1 are antiviral saponins yet can cause damage to red blood cells at effective drug concentrations that blocked viral entry (Fig 4). As such, Rev-1 and Gen-1 are both hemolytic, a property which may restrict potential use in therapy. To see whether modification of C28 sugar side chain can abolish such hemolytic effect, we acetylated the hydroxyl group of the C28 sugars with acetic anhydride. Briefly, 10ml of pyridine and 5ml acetic anhydride were added to a solution of pure tieghemella (Gen-1, 100 mg) in 5 ml methanol at 0° C. The solution mixture was allowed to reflux for five hours O 0 C. The resultant mixture was extracted with water at 2O 0 C as described in reference 9. The aqueous solution was washed several times with dichloromethane. The water filtrate was dried under the hood to afford oAc- Gen-1; (160mg). We compared the hemolytic effect of Gen-1 and o Ac-Gen- 1 by measuring the release of hemoglobin from red blood cells in the presence of Gen.1 or o-Ac Gen. The results demonstrated that modification of C28 sugars of Gen-1 by acetylation of the hydroxyl groups of the sugars can indeed eliminate the hemolytic effect of the Gen-1. As shown in figure 4, while

o-Ac-Gen-1 showed no hemolysis at 57μM, 50% of the red blood cells were lysed by Gen-1 at this concentration (9).

e. Acetylated Gen-1 is able to inhibit HIV-I replication with minimal effects on CD4 + cells.

A series of experiments demonstrated that the inhibitory effect of Gen 1 and acetyl Gen-1 on HIV attachment are similar, cells remained >95% viable above 90μM yet inhibited HIV replication (IC 90 ) at about 10 μM (Fig 5). Drug/virus Versus Drug/CD4 + cells preincubation studies have further indicated that acetyl Gen-1 targets HIV virus instead of host cells. For example, preincubation of o Ac-Gen- 1 with CD 4 + cells did not prevent HIV-I entry and infection in CD4 + cells while preincubation of o Ac-Gen- 1 with HIV-I virus totally inactivated the infectivity of the testing HIV-I (Fig 5, 6, Ref 9).

Example 2

III. Isolation of two highly potent and non-toxic inhibitors of HIV-I integrase from Salvia Miltiorrhiza. a. Salvia genus of Labiaceae family contains approximately 1000 species. 78 of these are distributed in different regions in China. Salvia miltiorrhiza root, commonly known as Dan- Shen root, has been used extensively for treatment of many diseases as antibacterial agent, for treatment of cardiovascular and cerebrovascular disease, against various types of hepatitis, chronic renal failure, etc. for centuries. Searches for chemical compounds which may be responsible for controlling these diseases has largely concentrated on using either lipophilic root extract or water soluble extract of the Dan-Shen roots. We found such a water soluble extract possesses exceptionally high antiviral activities against several types of viruses (HIV, SIV, HBS and MuLV) in infected cells and animals with extremely low acute toxicities (3). An IN 1 search was established based on an assay-guided purification procedure utilizing different concentrations of methanol, propanol, a two-layer extraction system (low salt water/butanol), and a high resolution inverted phase HPLC (Sperisorb OD S-2). From SM roots, two compounds (M 5 22 and M 5 32) were isolated from S. miltiorrhiza roots in purities of >99.5% as shown by NMR special analysis with yields of 0.018 and 0.38%, respectively (10). Structural determination revealed that M 5 22 is lithospermic acid and M 5 32 is lithospermic acid B (Fig 7). These two structurally related compounds are potent anti-HIV inhibitors and showed no cytotoxicity to H9 cells at high concentrations (CC100>297 μM for M 5 22 and >223 μM for M 5 32 (Fig 7). In addition, M 5 22 and M 5 32 inhibited HIV-I integrase catalytic activities of 3 '-joining to the target DNA with an IC50 of 0.48 μM for M 5 22 and 0.37 μM for M 5 32 (11). Compound binding to HIV-I integrase and inhibition of enzymatic activity within minutes. Neither M 5 22 nor M 5 32 by itself prevents

HIV entry in H9 cells. They also show no inhibition of reverse transcriptase activity in infected cells (10,16). The levels of intracellular strong stop and full-length viral DNA remained unchanged following drug treatment. However, both inhibitors strongly suppressed the acute HIV-I infection of H9 cells with IC50 values of 2 and 6.9 μM for M 5 22 and M 5 32, respectively. Thus these two selective integrase inhibitors should provide a novel class of therapeutic drugs for AIDS based on their high potencies and absence of cytotoxicity (10,11).

b. Inhibition of acute infection of different strains of HIV-I by M 5 22 and M 5 32

The inhibitory effect of M 5 22 and M 5 32 against HIV replication was further examined using 5 different strains of HIV-I, drug resistant and primary isolates in the presence of a variety of drug concentrations in infected H9 cells. By using the HIV/p24 monoclonal antibody assay, the IC 5O (50% inhibitory concentration) for M 5 22 against HIV-I drug resistant strain, HIV-IRTMF (AZT-resistant virus) was 2.5 μg/ml, 4.6 μM, HIV-1 π1B A17 variant (highly resistant to inhibition by RT nonnucleoside inhibitors) was 1.1 μg/ml, 2 μM and for HIV- 1 LIOR/M46I/L63P . V82T/I 84V (protease inhibitor resistant virus) the was 6 μg/ml, 11.1 μM. The HIV- 1 primary isolate strains had IC 50 5 S of 11 μg/ml, 20.4 μM for HIV- lm B and 9 μg/ml,

16.7 μM for HIV- 1 MN (Fig- 8A). Similar results were obtained for the inhibition effect of M 5 32 against replication in these five different strains of HIV-I. M 5 32 inhibited HγV1 RTM F (AZT-resistant virus) with an IC 5O of 6 μg/ml, 8.3 μM, HIV-lπ 1B A17 variant (highly resistant to inhibition by RT nonnucloside inhibitors) with an IC 50 of 5 μg/ml, 6.9 μM and HIV- 1 L I O R/M46 I/L63P /V 8 2T/I 84 V (protease inhibitor resistant virus) with an IC50 of 1 μg/ml, 1.3 μM while it inhibited the HIV-I primary isolate strains with an IC 5O of 7 μg/ml, 9.7 μM for HIV- lniB and of 6 μg/ml, 8.3 μM for HIV-1 MN (Fig. 8B).

c. M 5 22 and M 5 32 inhibit HIV integration step by targeting core domain of (A.A.50-212) of HIV-I integrase.

To determine whether M 5 22 and M 5 32 are acting against the catalytic core domain specifically, disintegration assay was tested using deletion mutant IN containing only the core domain of HIV integrase. Such deletion mutant is defective for normal integration, but it is capable of catalyzing a reversal of the forward reaction, termed "disintegration". The disintegration reaction can be assayed by using a deletion mutant HIV-I integrase (IN 50"212 ) which lacks both the amino terminal zinc binding and the carboxyl terminal DNA binding domains but remains active in this assay, whereas such truncated IN loses activity in the 3 '- processing and joining (strand transfer) assays. So, disintegration assay can be used to probe the site of drug-enzyme binding. As shown in Fig. 9, M 5 22 and M 5 32 inhibited both the wild type and the deletion mutant of HIV-I integrase (IN 50"212 ) with equal efficacy which implies that integrase core domain contains the binding site of M 5 22 and M 5 32 and is responsible for the HIV-I integrase inhibition (11).

d. M 5 22 and Ms32 protects infected cells from the cytopathic effects

A series of drug concentrations ranging from 1.25 μg/ml to 160 μg/ml were used for testing the cytotoxicity of M 5 22 and M 5 32 in H9 cells, using the MTT assay to determine the viability of H9 cells in culture. It was found that the cells remained viable during the entire culture period of 8 days even at the highest concentrations (CCi 00 ) of M 5 22 and M 5 32 tested (160 μg/ml, 297 μM for M 5 22 and 160 μg/ml, 223 μM for M 5 32. Inhibition of HIV-I replication by M 5 22 and M 5 32 was also examined in infected H9 cell cultures. H9 cells were infected with HIV-I RTMF (AZT-resistant virus) in the presence and absence of drugs. In the absence of drug syncytia are formed, while in the presence of drug, HIV-I replication was inhibited in H9 cells and syncytia formation was prevented. Protection of the cells from the cytopathic effects was observed. In fact, the drug-protected, HIV-infected H9 cells appeared to be morphologically identical to the uninfected control cells (6).

e. Interaction of Ms22 and Ms32 with amino acids in the catalytic core domain of HIV- lintegrase as determined by the docking calculations.

Although the binding modes of the two ligands are slightly different in some details, they shared some common binding sites (i) residues Glu92 and Lys 159 and (ii) the metal ion (11). Structural features of ligand that are crucial for the enzymatic activity are hydroxyl, aromatic ring, and carboxylic acid moieties. This could be supported by the docked conformations of both compounds M 5 22 and Ms32 showing that ligand metal interaction, hydrogen bond interaction and hydrophobic attraction played a critical role in the binding between HIV-I integrase and its inhibitors. This is described in more detail in reference 11.

Example 3

IV. NDGA Derivatives as Potent Mutation Insensitive Inhibitors for HSV, HPV and HIV proviral Transcription a. NDGA derivatives as transcription inhibitors for suppressing HIV and HSV viral growth.

At least 33 different plant lignans (derivatives of Nordihydroguaiaretic acid, NDGA have been found to possess cytostatic activities (12). Several lignans of species podphyllum are also found to be effective in treating of venereal warts, influenza A, vaccinia viruses, herpes simplex type I and II (13). At high concentrations, NDGA inhibits several enzymatic activities. This

harmful effect however can be greatly reduced when one of its catechol oxygens is blocked or when a hydrophobic group is added to the butane backbone in the mid-part of NDGA (14).

The mechanism underlying the antiviral action for NDGA derivatives was largely unknown before 1995. In 1995, Huang's group first reported a molecular study on 3'-O-methyl NDGA (Fig. 11, MaI 4)(15) isolated from creosote bush (Larrea tridentate) (16,3). We reported that 3'-0-methyl NDGA can specifically block HIV transcription, Tat-regulated transactivation, and HIV replication in human cell culture (3,15). NDGA derivatives interfere with the binding of transcription factor SpI to the promoter of the HIV proviral template. The target of Mal.4 is mapped to nucleotides -87 to -40, the SpI binding sites of the HIV long terminal repeat (LTR). Unmodified NDGA does not inhibit HIV transcription and has no effect on SpI binding in vitro. However, large preparations of such NDGA derivatives from plant sources are labor intensive and costly. In anticipation of the possible clinical use of lignans in controlling SpI -regulated viral and tumor growth in humans, nine different methylated NDGAs and several water soluble NDGA derivatives were synthesized chemically in large quantities with low cost (17, 18, 26, Fig. 11). Unsubstituted NDGA was used as the parent substrate. G 4 N ("tetraglycinal NDGA", see Fig 1, patent US 6,417,234Bl. Meso-l,4-BIS[3,4-(dimethylamino acetoxy)phenyl]-(2R,3S)- dimethyl butane hydrochloride salt was further used to conduct binding studies with HIV SpI enhancer sequence (3, 18, 19, 26, Fig 11). NDGA derivatives bind to the major groove of the free DNA when it is uncomplexed with other proteins. The binding constant for G 4 N/Spl enhancer sequence was calculated by the ethidium displacement technique at various phosphate buffer concentrations and pH values using a spectrofluorometer with excitation at 517nm and emission at 596nm. An apparent equilibrium binding constant (K app ) Of SxIO 6 M "1 was obtained by the equation K a pp=K EtBr [EtBr] / [G 4 N] which represents the concentration of the G 4 N causing a 50% reduction of the fluorescent intensity of DNA-ethidium solution (18). P 4 N, another water soluble NDGA derivative with four attached piperidino group, has been found to disrupt SpI transcription factor binding to the major groove of SpI site either by competition for binding or by altering the overall DNA conformation such that the major groove becomes incompatible for SpI binding (Fig. 11) (19, 26). The inhibitory effect of a variety of NDGA derivatives against transcription of a variety of viral genes of HIV, HPV, HSV and several oncogenes have been described in U.S. Pat. Nos. 5,663,209, 6,214,874, 6,291,524, 6,365,787, 6,417,234, and 6,608,108 and references 3, 15, 16, 17, 20, 21A, 21B, 23, 25.

b. NDGA Derivatives Are Potent Mutation-Insensitive Antiviral Agents against HSV with High Clinical and Therapeutic Advantages. The novel antiviral mode of action of NDGA derivatives confers several clinically relevant advantages which should significantly enhance their therapeutic value with regard to drug resistance, the prevention of HSV-I reactivation, and as topically applied agents for the treatment of herpes labialis. Because the inhibitory mechanism of this class of compounds is completely distinct from the antiviral mechanism of nucleoside analogue agents, there is little possibility of cross-resistance between these two classes of drugs. We have demonstrated that M 4 N (Fig. 12) retains complete efficacy against acyclovir-resistant strains of HSV-I (20), and the studies also showed that mutant acyclovir-resistant strains of HSV-I (HSV- 1 R ) have no cross-resistance against G 4 N (Figure 13) (21).

Another corollary to the mode of action of MaI.4, M 4 N and G 4 N is mutation-insensitivity. The capacity of harmful viral and bacterial strains in general, and of HSV-I specifically, to mutate in response to conventional chemical agents poses a rapidly increasing problem in new drug design and the development of therapeutic strategies. Viral inhibitors such as nucleoside analogues which target viral factors have the advantages of high specificity and low cytoxicity. However in the clinical setting, prolonged use of these drugs places selective pressure upon the viral population to generate harmful resistant strains containing mutant proteins against which the drug is ineffective. Mutant acyclovir-resistant strains of HSV-I have been commonly reported to contain altered TK and DNA polymerase enzymes (22), and repeated use of AZT and even the most effective reverse transcriptase (RT) and protease inhibitors leads to resistant HIV strains containing mutations found within HIV RT and protease enzymes. In contrast, host factors, unlike viral factors, are under no selective pressure to mutate and are in general structurally invariable. In comparative studies of mutation-insensitivity between M 4 N and acyclovir (20), there was no increase in the IC 50 OfM 4 N against HSV-I following ten passages of virus, indicating HSV-I was incapable of developing resistance to M 4 N over the short-term. In contrast, the IC 50 of acyclovir increased from 7.54 μM in the first passage to 444 μM in the tenth passage of HSV-I (Fig. 14, Ref. 21).

c. M 4 N and G 4 N as AntiHSV Agents in Animal Model Systems.

M 4 N and G 4 N were tested for their abilities to prevent HSV-I infection in two animal model systems. The dorsal cutaneous guinea pig model system is a well-established model for HSV-I infection that has been used in several studies to comparatively assess the potential efficacy of different topical formulations for treatment of herpes labialis. Straten MV, Carrasco D, Lee P, Tyring SK, Arch.Dermatol 137, 1232-1235 2001. Infected animals exhibited a dose-dependent response to treatment (measured by visual assessment): without G 4 N, HSV-I infection was obvious, at 5μM G 4 N, the drug was semi-effective and at 10μM, it was more effective and at 150μM G 4 N is extremely effective. G 4 N effectively inhibited HSV-I induced cutaneous lesion formation and viral shedding, with no evidence of toxicity or dermal irritation even at the highest soluble concentrations (150 mM).

The efficacies of M 4 N and G 4 N against HSV-2 in a mouse vaginal model system were also examined. Progestin-treated female CF-I mice (10 per group) were infected with HSV-2 in the absence or presence OfG 4 N (in PBS), P 4 N (in PBS), or M 4 N in Vaseline (23). At the end of three days, the vaginal lavages were collected from the washings of the vagina regions. The status of the vaginal HSV-2 infection and replication were examined by their cytopathic effects on cultured human foreskin fibroblast cells. Monitoring and scoring were carried out by three persons independently. Data indicated that with no drug treatment, 9 out of 10 animals were infected with an average score of 3.5 (>75% infection) and with G 4 N at O.lmM only 1 out of 10 was infected with an average score of 0.5 (app 12.5% infection). There were no infections detected with G 4 N treatment at 1OmM or 10OmM. P 4 N was also found to be quite effective. At 1OmM concentration only 2 of 10 mice were weakly infected. An average scale of 0.19 was obtained (Fig 15). For M 4 N treatment, because it was difficult to achieve total homogenous suspension OfM 4 N with Vaseline, data were less impressive. Out of 8 mice, 4 were fully infected with a score of 3-4. However, the other 4 showed very little infection at a M 4 N concentration of 20mg/ml. It is expected that better mixing OfM 4 N in Vaseline at a higher concentration than 20 mg/ml (55.8mM) should improve the efficacy of the treatment. All these compounds showed little toxicity in animals. For example, topical application for six days of G 4 N in guinea pigs at concentrations up to 50 mM showed no evidence of toxicity, and subdermal injection OfM 4 N also showed no toxicity in mice.

Example 4

Combinational Therapeutics Targeting Laboratory and Clinical Isolates of HIV-I.

Currently, there are 22 compounds representing four different mechanistic treatment classes that are used mostly in combination to treat HIV positive patients: nucleoside and nonnucleoside inhibitors for virus transcriptase (NRTi and NNRTi), viral protease inhibitors and viral fusion inhibitors. Combinational therapies using drugs from these different mechanistic classes were initially quite effective, which have resulted in overall decrease of HIV-I morbidity and mortality in recent years. However, emergence of multidrug-resistent strains, and treatment related host toxicities became increasingly common with long time users of such drug treatment regimens. The discovery of host membrane protein CCR5 as coreceptor essential for CD4/gpl20 interaction and viral-cellular fusion has led to the findings of CCR5 antagonists, MARA VIROC, Vicritrinoc, TAK-779 and the development of monoclonal antibody PRO140 against CCR5 recently. PRO 140 binds as a competitive inhibitor for extracellular epitope on CCR5, while small molecular antagonists target hydrophobic pocket of CCR5. Antiviral synergy was reported between PRO 140 and small-molecule CCR5 antagonists and with other classes of HIV inhibitors using HIV-I pseudovirus assay, a self-inactivating vector from pNL4- 3δENV-leuciferase vector (24).

Our combinational regimen is unique in several ways. (1) We have identified two HIV integrase inhibitors, M 5 22 and M 5 32, that are exceedingly active and non-toxic. (2) We have isolated a small molecular weight viral entry inhibitor (Gen-1) which uses its carbohydrate moiety to block HIV gpl20 fusion, a mechanism different from others reported. (3) We have identified another mechanistic class inhibitor which blocks proviral transcription and post-integrations (for example, G 4 N, Fig 11) and is equally active toward both wild type and drug resistant strains of HIV-I. This class of viral inhibitor targets viral transcription which uses all host cellular machinery that is not subject to selective pressure to mutate from drug treatment as shown for other viral protein inhibitors. We have demonstrated that when used in combination these compounds have a synergistic effect that permits the use of much lower dosages than would be otherwise necessary to achieve a therapeutic effect.

The efficacies of these drugs were tested individually and in combination against infection and replication of many strains of HIV-I including the fresh clinical isolates from the infected patients.

We tested drug combinations against two different cells infected with laboratory strain of HIV-I RTMF. For these drug combinational studies, both the H9 cells and human PBMC have been used. We first tested how our inhibitors affect one specific strain of HIV which is resistant to drug against RT (HIV-I RTMF )- Briefly H9 (lxlO 5 /ml) were infected with HIV-I RTMF strain with a multiplicity of infection of 0.1. These cells were further tested with three drugs, M 5 22, Gen-1 and G 4 N, either in combination or separately (8,10,21B). After 4-day incubation at 37°C, H9 cells were subcultured with fresh culture medium containing appropriate concentrations 0, 1.25, 2.5, 5, 10, 20, 40 and 80 μM of each of the drugs, or of a combination of all three of the drugs, each at the indicated concentration. The inhibition for HIV-I by these combinations of drugs was examined using p24 assay after 8 days. It was found that IC 50 for the combination of the three drugs against HIV-I is 1.3 μM (Fig 16). In other words as combinational therapy, a IC 50 of 1.3μM was achieved when 1.3μM of each of these three drugs was used in combination..

Similarly, PBMC cells were isolated from a healthy blood sample and these cells were stimulated by PHA and IL-2. PBMC (5x10 6 /ml) were infected with HIV-I RTMF - The cells were treated with three combinations of drugs, M 5 22, Gen-1 and G 4 N. Fresh media and drugs were added after 4 days with drug concentrations 0, 1.25, 2.5, 5, 10, 20, 40 and 80 μM. The inhibition for HIV-I by these combinations of drugs was also examined by using p24 assay after 8 days. It was found that IC 50 for the combinations of three drugs against HIV-I is 1.25 μM. On the other hand when tested alone with HIV-I RTMF in H9 cells the IC 50 for Gen-1 (viral entry inhibitor), M 5 22 the integration inhibitor and G 4 N, proviral transcription inhibitor, are 40 μM, 4-6 μM and 18 μM respectively (See Fig. 16-18). In comparison, cytotoxicity for the three drugs combined to H9 cells was also analyzed using an MTT assay. As shown in Fig 18, cellular toxicity to H9 cells combined was not detected in the presence of the three drugs, even when 12 μM of each was used.

In addition, the three drugs are being tested individually and in combination on PBMC cells infected with fresh clinical isolates of HIV-I from ADDS patient. Blood samples were collected

from a healthy donor, and PBMC cells were isolated by Ficoll-Hypaque density gradient centrifugation. These cells were stimulated by PHA and IL-2. PBMC cells (5xlO 6 /ml), and infected with the clinical isolates of HIV-I from an infected ADDS patient. These cells will be treated with the three drugs individually and in combination of M522, Gen-1 and G4N. Fresh media and drugs will be added every 4 days with drug concentrations 0, 1.25, 2.5, 5, 10, 20, 40 and 80 μM. The inhibition of HIV-I by these drugs individually and in combinations will be tested by using p24 assay after 8 days similar to the protocol shown in figure 19. The results are expected to further confirm the advantages of the combination therapy.

References cited herein are listed below for convenience and are hereby incorporated by reference.

1. Altman LK "AIDS is on the rise worldwide, U.N. Finds New York Times, Nov. 22, 2006 www. UNAEDS

2. Gallo RC. The end or the beginning of the drive to an HIV-preventive vaccine: A view from over 20 years. 3. Huang RCC, Chang CC, Mold D. Survivin dependent and independent pathways and the induction of cancer cell death by tetra-O-methyl nordihydroguaiaretic acid. Editors D. Von Hoff and Gray P, Invited Article, Seminars in Oncolosv 33 (4), 479-485 (2006).

4. Gosse BK, Gnabre JN, Ito Y, Huang RC. Isolation of saponins with viral entry inhibitory activity by combined chromatographic methods. J Liq Chromatosr ReI Technol., 25: 3197-

3209 (2002).

5. Gosse BK, Gnabre JN, Bates RB, Nakkiew P, Huang RCC. Antiviral saponins from Tieghemella heckelii. J Nat Products. 65, 1942-1944 (2002).

6. Abd-Elazem IS. Isolation of two highly potent and non toxic inhibitors of human immunodefficiency virus type I (HIV-I) integrase and viral replication. JHU Ph.D. Thesis 2002. 7. Este JA, Cabrera C, Schols D, Cherepanov P, Gutierrez A, Witrouw M, Pannecouque C, Debyser CZ, Rando RF, Clotet B, Desmyter J, and DeClerco E. Human immunodeficiency virus glycoprotein gpl20 as the primary target for antiviral action of AR177 (Vintevir). MoI Pharmacol 53, 340-345 (1998). 8. Gnabre J, Gosse B, Park R, Gallay P, Abd-Elazem IS and Huang RC. Triterpenoid saponins inhibit HSV-I and HIV-I infections by blocking viral entry. Manuscript in preparation.

9. Gosse B, Abd-Elazem IS and Huang RC. Acetylation of C28 sugars of GEN-I abolishes antiviral activities without cellular toxicity. Manuscript in preparation.

10. Abd-Elazem IS, Chen HS, Bates RB, Huang RCC. Isolation of two highly potent and nontoxic inhibitors of human immunodeficiency virus type 1 (HIV-I) integrase from Salvia miltiorrhiza. Antiviral Res.. 55, 91-106 (2002).

11. Abd-Elazem IS, Nunthaboot N, Kokpol S, Painwanit S, and Huang RC. Inhibition of replication of drug resistant and primary isolates of HIV-I by specific integrase inhibitors, M522 and M532. Manuscript in preparation for submission to J Virol. 12. Dewick PM and Jackson DE. Cytotoxic lignans from popodhylum and the nomenclature of aryltetralin lignans. Pytochemistry 20, 2277 (1981).

13. Bedows E and Hatfield GM. Studies on Podophyllotoxin, β-peltatin,

Deoxypodophyllotoxin Picropodophyllotoxin and α-peltatin against Measles and Herpes Simplex Type I. J. Nat Prod 45, 725 (1978). 14. Schegg KM and Welch W. the effect of Nordihydroguaiaretic Acid and related lignans on Formyltetra Hydrofolate Synthesis and Carboxyl-Esterase. Biochem Biophys ACTA 788, 167 (1984).

15. Gnabre JN, Brady JN, Clanton DJ, Ito Y, Dittmer J, Bates RB, and Huang RC. Inhibition of human immunodeficiency virus type I transcription and replication by DNA sequence- selective lignans. Proc Natl Acad Sci USA 92, 11239-11243 (1995).

16. Gnabre, JN, Bates, RB, Caldera, S and Huang, RC. Chemical Structure of HIV-Inhibitory Lignans from Larrea tridentata. Tetrahedron, 51, 12203-12210, 1995.

17. Hwu JR, Tseng WN, Gnabre J, Giza P, Huang RC. Antiviral activities of methylated nordihydroguaiaretic acids (I) Synthesis, structure identification and inhibition of Tat regulated HIV transactivation. J Medicinal Chem., 41, 2994-3000 (1998). 18. King KY, Hakimelahi GH, Huang RC and Hwa JR. Design, synthesis and binding studies of highly water soluble Nordihydroguaiaretic acid derivatives: a DNA major groove binder. J of Gen and MoI Biol 13 (No. 4), 248-257 (2002).

19. Dohm JA, Hsu M-H, Hwu J-R, Huang RCC, Moudrianakis EN, Lattman EE, Gittis AG. Influence of polyamines, ions, hydration, and the transcriptional inhibitor P4N on the conformations of the SpI binding site. J Molec Bio 349, 731-744, (2005).

20. Chen H, Li T, Li JN, Park R, Mold D, Gnabre J, Hwu JR, Tseng WN, Huang RCC. Antiviral activities of methylated nordihydroguaiaretic acids (II) Targeting herpes simplex virus replication by mutation insensitive transcription inhibitor terra- O-methyl NDGA. J

Medicinal Chem., 41, 3001-3007 (1998).

21 A. Park R, Giza PE, Mold DE, Huang RCC. Inhibition of HSV-I replication and reactivation by the mutation-insensitive transcription inhibitor tetra-O-glycyl-nordihydroguaiaretic acid. Antiviral Res., 58, 35-45 (2003).

2 IB. Huang RCC, Li Y, Giza PE, Gnabre JN, Abd-Elazem IS, King KY, Hwu JR. Novel antiviral agent tetraglycylated nordihydroguaiaretic acid hydrochloride as a host-dependent viral inhibitor. Antiviral Res.. 58, 57-64 (2003).

22. Hirsch MD, Kaplan JC, D'Aquila RI. Raven, Philadelphia. Antiviral Agents. In: Fields

BN, Knipe DM, Howley PM, Chanock RM, Melnick JC, Monath TP, et al. (Eds.), Fields

Virology, 3 rd ed pp. 431-466, 1996. 23. Park R, Mold DE and Huang RC. Direct virucidal effect of tetra-o-glycyl- nordihydroguaiaretic acid (G4N) on HSV-I infection of vero cells and in a dorsal cutaneous guinea pig model system. Manuscript in preparation.

24. Murga JO, Franti M, Pevear PJ, Maddon PJ and Olson WC. Potent antiviral synergy between monoclonal antibody and small-molecule CCR5 inhibitors of human immunodeficiency virus type I. Antimocrobial Agents and Chemotherapy vol. 50, No. 10 p3289-3296 (2006)

25. Craigo J, Callahan M, Huang RC, Delucia AL. Inhibition of human papilloma virus type 16 gene expression by nordihydroguaiaretic acid plant lignan derivatives. Antiviral Res.. 47, 19-28 (2000).

26. Hwu JR, Hsu M-H, Gitti R, Huang RCC. New nordihydroguaiaretic acid derivatives as anti-HFV agents. J Med Chem, Manuscript submitted.